1
|
Lee CC, Tu YC, Wu HT, Ko WC, Liu HC, Tsai PJ, Chang HN, Huang IH, Hung YP. Clostridium Butyricum Miyairi Bacteriocin Treatment for Clostridioides difficile Infections with Clinical Isolates: Insights from In Vitro, Ex Vivo, and Mouse Model Studies. J Glob Antimicrob Resist 2025:S2213-7165(25)00066-9. [PMID: 40147758 DOI: 10.1016/j.jgar.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVE The standard antimicrobial therapy for Clostridioides difficile infections (CDIs) is limited to oral fidaxomicin or vancomycin, but these agents are associated with high treatment failure and recurrence rates. Clostridium butyricum had been proven effective in many kinds of gastrointestinal disease. With a less disturbed gut microbiota, we hypothesized that the properties of Clostridium butyricum Miyairi Bacteriocin (CBM-B) make it a potential therapeutic agent for treating patients with CDIs. METHODS The inhibitory effects of CBM-B and vancomycin were compared using the kinetic time-kill assay, ex vivo co-culture model and mouse model. RESULTS Among the clinical isolates of C. difficile, the minimal inhibitory concentration (MIC) of CBM-B ranged from 0.0625 to 8 µg/ml; the MIC50 and MIC90 were 1 µg/ml and 4 µg/ml, respectively. In the mouse model infected with a RT078 and receiving CBM-B intra-rectal enema therapy, mice infected with isolates with a relative low CBM-B MICs (2 µg/ml, abbreviated as M2) revealed significant better therapeutic effect, including less loss of body weight and cecum weight, compared with those infected with isolates of relative high CBM-B MICs (4 or 8 µg/ml, abbreviated as M4 or M8) . The relative C. difficile bacterial burden in stool of mice receiving CBM-B treatment were significantly lower among mice infected with M2, compared with that infected with M4 or M8. CBMB treatment, compared with vancomycin therapy revealed less disturbance in gut microbiota. CONCLUSION CBM-B could be effective in the treatment of CDIs that infected with a C. difficile isolate with relatively low MICs with less disturbance in gut microbiota.
Collapse
Affiliation(s)
- Ching-Chi Lee
- Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chen Tu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Tsung Wu
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Chieh Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| | - Pei-Jane Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiang-Ning Chang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Hsiu Huang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences.
| | - Yuan-Pin Hung
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan.
| |
Collapse
|
2
|
Mears KS, Denny JE, Maslanka JR, Mdluli NV, Hulit EN, Matsuda R, Furth EE, Buffie CG, Abt MC. Therapeutic activation of IL-22-producing innate lymphoid cells enhances host defenses to Clostridioides difficile infection. Cell Rep 2025; 44:115438. [PMID: 40138315 DOI: 10.1016/j.celrep.2025.115438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/02/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Clostridioides difficile causes debilitating colitis via secreted toxins that disrupt the intestinal barrier, and toxemia is associated with severe disease. Thus, therapies that fortify the intestinal barrier will reduce the severity of infection. Innate lymphoid cells (ILCs) are critical in the defense against acute C. difficile infection and represent a promising therapeutic target to limit disease. Here, we report that oral administration of the Toll-like receptor (TLR) 7 agonist R848 limits intestinal damage and protects mice from lethal C. difficile infection without impacting pathogen burden or altering the intestinal microbiome. R848 induced interleukin (IL)-22 secretion by ILCs, leading to STAT3 phosphorylation in the intestinal epithelium and increased stem cell proliferation. Genetic ablation of ILCs, IL-22, or epithelial-specific STAT3 abrogated R848-mediated protection. R848 reduced intestinal permeability following infection and limited systemic toxin dissemination. Combined, these data identify an immunostimulatory molecule that activates IL-22 production in ILCs to enhance host tissue defenses following C. difficile infection.
Collapse
Affiliation(s)
- Kevin S Mears
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua E Denny
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey R Maslanka
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nontokozo V Mdluli
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellie N Hulit
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rina Matsuda
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Emma E Furth
- Department of Pathology, University of Pennsylvania Medical Center, Philadelphia, PA, USA
| | - Charlie G Buffie
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, New York, NY, USA
| | - Michael C Abt
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Zhang T, Villalba MI, Gao R, Kasas S, von Gunten U. Effect of surfactants on inactivation of Bacillus subtilis spores by chlorine. WATER RESEARCH 2025; 272:122944. [PMID: 39708383 DOI: 10.1016/j.watres.2024.122944] [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: 09/16/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024]
Abstract
Bacterial spores pose significant risks to human health, yet the inactivation of spores is challenging due to their unique structures and chemical compositions. This study investigated the synergistic effect between surfactants and chlorine on the inactivation kinetics of Bacillus subtilis spores. Two surfactants, cocamidopropyl betaine (CAPB) and cetyltrimethylammonium chloride (CTMA) were selected to investigate chlorine disinfection in absence and presence of surfactants. The concurrent presence of both chlorine and surfactant resulted in a moderate reduction in the lag-phases for spore inactivation and negligible increase in the second-order inactivation rate constants. In contrast, when the spores were pre-exposed to surfactants, the lag-phases decreased by about 50 % for both CAPB and CTMA, and the second-order inactivation rate constants during post-chlorination remained constant for CAPB but increased by a factor of 2.3 for CTMA, compared to the control group with phosphate buffer. This synergistic effect became more pronounced with longer surfactant pre-exposure times, reaching its maximum at 3-6 h. The observed synergistic effect suggests that surfactants can potentially enhance the permeability of the coat which is the outmost layer of B. subtilis spores and a primary barrier for chemical disinfectants. Tracing a group of B. subtilis spores sequentially treated with surfactant and chlorine by atomic force microscopy, a significant decrease in compressive stiffness of the spores was observed due to exposure to surfactants, indicating alterations in the coat by surfactants. The trend in reducing compressive stiffness aligned well with the decrease of lag-phases in inactivation kinetics. Furthermore, CTMA was found to inactivate B. subtilis spores through mechanisms different from chlorine. Chlorine primarily inactivated B. subtilis spores before damaging the inner membrane of the spores which plays a crucial role in protecting the genetic material stored in the core of the spores. In comparison, CTMA damaged 22 % of the inner membrane for an inactivation efficiency of 99 %. A synergistic effect in damaging the inner membrane was observed when applying CTMA and chlorine simultaneously instead of sequentially.
Collapse
Affiliation(s)
- Tianqi Zhang
- School of Architecture, Civil and Environmental Engineering (ENAC), École Polytechnique Fedérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - María Inés Villalba
- Laboratory of Biological Electron Microscopy (LBEM), École Polytechnique Fédérale de Lausanne (EPFL) and Université de Lausanne (UNIL), CH-1015 Lausanne, Switzerland
| | - Rongjun Gao
- School of Architecture, Civil and Environmental Engineering (ENAC), École Polytechnique Fedérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Sandor Kasas
- Laboratory of Biological Electron Microscopy (LBEM), École Polytechnique Fédérale de Lausanne (EPFL) and Université de Lausanne (UNIL), CH-1015 Lausanne, Switzerland; Centre Universitaire Romand de Médecine Légale, UFAM, Université de Lausanne, 1015 Lausanne, Switzerland
| | - Urs von Gunten
- School of Architecture, Civil and Environmental Engineering (ENAC), École Polytechnique Fedérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland; Eawag, Swiss Federal Institute of Aquatic Science and Technology (Eawag), CH-8600 Dübendorf, Switzerland.
| |
Collapse
|
4
|
Watanabe K, Tsuji T, Matsuzawa H, Saruta Y, Shimodaira Y, Iijima K. A Simple Prediction Model for Clostridioides difficile Infection: A Hospital-Based Administrative Database Study. J Gastroenterol Hepatol 2025; 40:609-617. [PMID: 39690954 DOI: 10.1111/jgh.16851] [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: 07/29/2024] [Revised: 10/30/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND AND AIM Few prediction scores for Clostridioides difficile infection (CDI), a potentially life-threatening nosocomial diarrhea, combine high accuracy with simplicity. A simple prediction score for routine clinical practice is needed. METHODS We conducted a retrospective cohort study of all inpatients aged ≥ 18 at a secondary care hospital in Japan. The derivation and validation cohorts consisted of patients from January 2016 to December 2020 and January 2021 to September 2022, respectively. Demographic and clinical data were retrieved using electronic medical records and an administrative database. The primary outcome was to derive and validate an accurate, simple prediction score for primary hospital-onset CDI. A derived prediction score by logistic regression analysis was calibrated and validated. RESULTS CDI developed in 102 of 25 517 and 25 of 6259 patients in the derived and validation cohorts (2.7 cases/10 000 patient-days). The derived model for predicting CDI, including antibiotic use, acid suppressant (proton pump inhibitors or vonoprazan) use, Charlson comorbidity index, and Barthel index, yielded c-statistics of 0.89 and 0.82 in the derivation and validation cohort. The model was well calibrated. CONCLUSIONS This simple prediction score enables early medical intervention and modification of treatment plans to reduce the risk of developing primary hospital-onset CDI.
Collapse
Affiliation(s)
- Kenta Watanabe
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Tsuyotoshi Tsuji
- Department of Gastroenterology, Akita City Hospital, Akita, Japan
| | | | - Yohei Saruta
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yosuke Shimodaira
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Katsunori Iijima
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
5
|
Naushad VA, Purayil NK, Varikkodan I, Badi AM, Baghi MA, Chandra P, Alarbi KMS, Althani MK, Aboughalia AT, Farooqi A, Kartha AB, Elzouki A. Demographic characteristics and clinical and laboratory features of patients with Clostridiodes difficile infection: A retrospective study in Qatar. IJID REGIONS 2025; 14:100592. [PMID: 40114911 PMCID: PMC11923805 DOI: 10.1016/j.ijregi.2025.100592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 03/22/2025]
Abstract
Objectives Clostridioides difficile infection (CDI) is one of the most common causes of diarrhea in healthcare facilities, imposing a significant burden on health-related budgets and resources worldwide. We aimed to study the demographic features, laboratory findings, and outcomes of CDI in Qatar. Methods A retrospective study involving adult patients with a confirmed diagnosis of CDI was conducted. Results Of the 595 patients, 308 (51.8%) were men with a mean age of 58 ± 19.9 years. The median duration of symptoms was 2 days, with an interquartile range values of 2-4 days. The most common symptoms reported were diarrhea (90.6%) and abdominal pain (41.5%). A total of 426 (71.6%) and 422 (70.9%) patients had a history of exposure to antibiotics and proton pump inhibitors, respectively, while 461 (77.7%) patients had a history of contact with a healthcare facility in the prior 3 months. Respiratory tract infections (33.9%) and urinary tract infections (22.4%) were the most common indications for antibiotic use in the study population. Increased C-reactive protein levels were the most frequently observed laboratory findings. Sepsis was identified as the most common complication (10.4 %). Reinfection was observed in 75 (12.6%) patients. Vancomycin monotherapy was the most commonly prescribed treatment. Conclusions In Qatar, CDI remains a significant health concern, primarily affecting elderly men, especially those who have had hospital admissions or used proton pump inhibitors or antibiotics. Preventive measures and increased knowledge of contact precautions and hand hygiene, particularly among healthcare workers, will help to reduce transmission. Raising physician awareness regarding the prudent use of antibiotics and antibiotic stewardship will serve as an adjunct to reduce the incidence of CDIs.
Collapse
Affiliation(s)
- Vamanjore A Naushad
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
- Department of Clinical Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Nishan K Purayil
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
- Department of Clinical Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Irfan Varikkodan
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
| | - Ahmed M Badi
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
| | - Mohamed A Baghi
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
- Department of Clinical Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Prem Chandra
- Medical Research Center, Hamad Medical Corporation, Doha, Qatar
| | - Khaled M S Alarbi
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Maryam K Althani
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
| | - Ahmed T Aboughalia
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
| | - Amer Farooqi
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Anand B Kartha
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
- Department of Clinical Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Abdelnaser Elzouki
- Department of Medicine, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Clinical Department, College of Medicine-QU Health, Qatar University, Doha, Qatar
- Department of Clinical Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| |
Collapse
|
6
|
Tang M, Wu Y, Olnood CG, Gao Y, Wang F, Zhang Z, Peng C, Zhou X, Huang C, Xiong X, Yin Y. Effects of peroxidized lipids on intestinal morphology, antioxidant capacity and gut microbiome in piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:430-443. [PMID: 40034456 PMCID: PMC11875184 DOI: 10.1016/j.aninu.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025]
Abstract
This study investigated the effect of peroxidized lipids on piglets' growth performance, intestinal morphology, inflammatory reactions, oxidative stress in the liver, duodenum, jejunum, ileum, and colon, and ileal microbiota. Twenty piglets (Duroc × [Landrace × Yorkshire]; age = 21 d old, BW = 6.5 ± 1 kg) were randomly assigned to two groups with 10 replicates per group and one piglet per replicate. The control group was fed 6% fresh soybean oil and the peroxidized soybean oil (PSO) group fed 6% PSO. The experimental feeding period lasted 24 d. The study found no impact on ADFI, ADG and gain to feed ratio (P > 0.05). However, the PSO group increased the diarrhea index and the serum levels of lactate dehydrogenase triglycerides, cholesterol, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol (P < 0.05), along with decreased concentrations of alanine aminotransferase and blood urea nitrogen (P < 0.05). For oxidative enzymes, PSO increased the concentration of F2-isoprostane in urine (P = 0.032), malondialdehyde (MDA) in the duodenum (P = 0.001) and jejunum (P = 0.004), decreased thiobarbituric acid reactive substances (TBARS) in the liver (P = 0.001) but increased TBARS in duodenum (P = 0.001), and carbonylated proteins in the duodenum (P = 0.003). For antioxidant enzymes, PSO decreased superoxide dismutase (SOD) in the liver (P = 0.001), colon (P = 0.002), and jejunum (P = 0.015), along with glutathione peroxidase (GSH-Px) in the liver (P = 0.008) and NAD(P)H:quinone oxidoreductase 1 (NQO1) in ileum (P = 0.001). For inflammatory reactions, PSO increased interleukin (IL)-1β concentrations in the duodenum and colon, and IL-10 in the jejunum, while decreasing IL-4 concentration in the duodenum (P < 0.05). For intestinal morphology and ileal microbiota, PSO increased ileal crypt depth, while decreasing the crypt-to-villus ratio (P < 0.05). Peroxidized soybean oil increased the relative abundance of Prevotella, Clostridium_sensu_stricto_1, Clostridium_sensu_stricto_6, Pasteurella and Klebsiella (P < 0.05). In conclusion, this study revealed that PSO worsened diarrhea, increasing the ileal crypt depth and the relative abundance of harmful microbiota, and induced oxidative stress and inflammation in the intestines and liver, primarily in the jejunum and ileum.
Collapse
Affiliation(s)
- Mengxuan Tang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yuliang Wu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | | | - Yundi Gao
- Sichuan Synlight Biotech Ltd., Chengdu 610041, China
| | - Fei Wang
- Sichuan Synlight Biotech Ltd., Chengdu 610041, China
| | - Zicheng Zhang
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Can Peng
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | - Xihong Zhou
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| | - Chunxia Huang
- School of Stomatology, Changsha Medical University, Changsha 410219, China
| | - Xia Xiong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
- School of Stomatology, Changsha Medical University, Changsha 410219, China
| | - Yulong Yin
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Chinese Academy of Sciences, Changsha 410125, China
| |
Collapse
|
7
|
Hirsch W, Fischer M, Khoruts A, Allegretti JR, Kelly CR, Vaughn B. Risk Factors for Antibiotic Exposure Post-Fecal Microbiota Transplantation for Recurrent Clostridioides difficile Infection: A Prospective Multicenter Observational Study. Open Forum Infect Dis 2025; 12:ofaf130. [PMID: 40103733 PMCID: PMC11913780 DOI: 10.1093/ofid/ofaf130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Background Recurrent Clostridioides difficile infection (CDI) is primarily driven by antibiotic-induced disruption of the indigenous intestinal microbiota. Restoration of microbiota through fecal microbiota transplantation (FMT) is effective in preventing subsequent CDI, although this effect is attenuated with additional antibiotic exposure. The aim of this study was to identify the risk factors for recurrent antibiotic administration after FMT. Methods This is a prospective cohort of patients who were administered FMT for recurrent CDI from 1 July 2019 through 23 November 2023 across 6 institutions in the United States. Providers collected de-identified data at the time of FMT administration and in the months post-FMT administration. Results The analysis included 448 patients. Risk factors for non-CDI antibiotic administration within 2 months of FMT included immunocompromised status (odds ratio [OR], 2.2 [95% confidence interval {CI}, 1.1-4.4]; P = .02), >3 non-CDI antibiotic courses pre-FMT (OR, 3.1 [95% CI, 1.4-6.8]; P = .006), and prior hospitalization for CDI (OR, 2.0 [95% CI, 1.1-3.8]; P = .02). The most common indications for non-CDI antibiotic administration post-FMT were urinary tract infections, respiratory infections, and procedure prophylaxis. Conclusions Non-CDI antibiotic exposure significantly increases the risk of CDI recurrence post-FMT. Risk factors for non-CDI antibiotic administration within 2 months of FMT include immunocompromised status, multiple prior non-CDI antibiotics, and prior hospitalization for CDI. These individuals may benefit from additional or modified recurrent CDI prevention strategies.
Collapse
Affiliation(s)
- William Hirsch
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Monika Fischer
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- BioTechnology Institute, University of Minnesota, St Paul, Minnesota, USA
| | | | - Colleen R Kelly
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Byron Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
8
|
Wu B, Song X, Liu Y, Zheng X. Clostridium difficile Bacteremia in an Elderly Patient with Multiple Comorbidities: A Case Report. Surg Infect (Larchmt) 2025; 26:112-115. [PMID: 39605192 DOI: 10.1089/sur.2024.223] [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: 11/29/2024] Open
Abstract
Clostridium difficile (C. difficile) stands as a primary cause of health-care-associated colitis in adults; however, extraintestinal manifestations of C. difficile, particularly bacteremia, are exceptionally rare. In this report, we document a case of an elderly male with multiple comorbidities who presented with an acute onset of fever. Diagnostic testing revealed the presence of concurrent bacteremia involving C. difficile and Klebsiella pneumonia. The multilocus sequence typing analysis identified this C. difficile strain as ST81. After receiving a combination treatment of vancomycin and biapenem, the patient successfully recovered and was subsequently discharged. This case report elucidates the clinical presentation and treatment strategies for C. difficile ST81 bacteremia, underscoring the critical need for heightened monitoring of extraintestinal infections in high-risk patients.
Collapse
Affiliation(s)
- Binwei Wu
- Qingdao Municipal Hospital, Qingdao, China
| | | | - Yu Liu
- College of Geodesy and Geomatics, Shandong University of Science and Technology, Qingdao, China
| | - Xu Zheng
- Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
9
|
Kuziel GA, Lozano GL, Simian C, Li L, Manion J, Stephen-Victor E, Chatila T, Dong M, Weng JK, Rakoff-Nahoum S. Functional diversification of dietary plant small molecules by the gut microbiome. Cell 2025:S0092-8674(25)00155-2. [PMID: 40056901 DOI: 10.1016/j.cell.2025.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/09/2024] [Accepted: 01/31/2025] [Indexed: 03/10/2025]
Abstract
Plants are composed of diverse secondary metabolites (PSMs), which are widely associated with human health. Whether and how the gut microbiome mediates such impacts of PSMs is poorly understood. Here, we show that discrete dietary and medicinal phenolic glycosides, abundant health-associated PSMs, are utilized by distinct members of the human gut microbiome. Within the Bacteroides, the predominant gram-negative bacteria of the Western human gut, we reveal a specialized multi-enzyme system dedicated to the processing of distinct glycosides based on structural differences in phenolic moieties. This Bacteroides metabolic system liberates chemically distinct aglycones with diverse biological functions, such as colonization resistance against the gut pathogen Clostridioides difficile via anti-microbial activation of polydatin to the stilbene resveratrol and intestinal homeostasis via activation of salicin to the immunoregulatory aglycone saligenin. Together, our results demonstrate generation of biological diversity of phenolic aglycone "effector" functions by a distinct gut-microbiome-encoded PSM-processing system.
Collapse
Affiliation(s)
- Gavin A Kuziel
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriel L Lozano
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Corina Simian
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology & Department of Bioengineering, Northeastern University, Boston, MA 02120, USA; Institute for Plant-Human Interface, Northeastern University, Boston, MA 02120, USA
| | - Long Li
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - John Manion
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Emmanuel Stephen-Victor
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Talal Chatila
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Jing-Ke Weng
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology & Department of Bioengineering, Northeastern University, Boston, MA 02120, USA; Institute for Plant-Human Interface, Northeastern University, Boston, MA 02120, USA
| | - Seth Rakoff-Nahoum
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Hasan MK, Alaribe O, Govind R. Regulatory networks: Linking toxin production and sporulation in Clostridioides difficile. Anaerobe 2025; 91:102920. [PMID: 39521117 PMCID: PMC11811957 DOI: 10.1016/j.anaerobe.2024.102920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Clostridioides difficile has been recognized as an important nosocomial pathogen that causes diarrheal disease as a consequence of antibiotic exposure and costs the healthcare system billions of dollars every year. C. difficile enters the host gut as dormant spores, germinates into vegetative cells, colonizes the gut, and produces toxins TcdA and/or TcdB, leading to diarrhea and inflammation. Spores are the primary transmission vehicle, while the toxins A and B directly contribute to the disease. Thus, toxin production and sporulation are the key traits that determine the success of C. difficile as a pathogen. Both toxins and spores are produced during the late stationary phase in response to various stimuli. This review provides a comprehensive analysis of the current knowledge on the molecular mechanisms, highlighting the regulatory pathways that interconnect toxin gene expression and sporulation in C. difficile. The roles of carbohydrates, amino acids and other nutrients and signals, in modulating these virulence traits through global regulatory networks are discussed. Understanding the links within the gene regulatory network is crucial for developing effective therapeutic strategies against C. difficile infections, potentially leading to targeted interventions that disrupt the co-regulation of toxin production and sporulation.
Collapse
Affiliation(s)
- Md Kamrul Hasan
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Oluchi Alaribe
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Revathi Govind
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
13
|
Hsia CH, Su HY, Chien YW. Risk Factors for Clostridium difficile Infection in Inpatients: A Four-Year (2017-2020) Retrospective Study. Antibiotics (Basel) 2025; 14:133. [PMID: 40001377 PMCID: PMC11851458 DOI: 10.3390/antibiotics14020133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Clostridium difficile infection (CDI) impact on healthcare systems is concerning due to high treatment cost and increased hospitalization time. We found that the incidence rate of CDI increased annually at Taipei Medical University Hospital (TMUH). The study aimed to establish monitoring indicators for hospitalized patients receiving antibiotics to prevent CDI occurrences. METHODS A case-control study was conducted to identify the risk factors of CDI among patients who were admitted to TMUH and tested for C. difficile. Patient demographics, patient history, and laboratory data were collected and analyzed. RESULTS Nutrition Risk Screening 2002 score (NRS 2002) in CDI patients was significantly lower than in non-CDI patients (3.1 ± 1.7 vs. 3.5 ± 1.6). The percentage of tube feeding in CDI patients was significantly lower than in non-CDI patients (23.0% vs. 36.7%), whereas parenteral nutrition was higher (8.8% vs. 3.8%). Age (OR = 1.03, p = 0.00), NRS 2002 score (OR =0.86, p = 0.05), comorbidity with cardiovascular disease (OR = 1.73, p = 0.03) or pulmonary disease (OR = 3.72, p = 0.00), patients with tube feeding (OR = 2.02, p = 0.01), and the number of medications (OR = 1.15, p < 0.01) were significant predictors of CDI. CONCLUSION This study revealed that nutritional factors, including NRS 2002 scores and feeding routes, were associated with CDI, emphasizing the importance of nutritional factors as key predictors in managing and preventing CDI.
Collapse
Affiliation(s)
- Chu-Hsuan Hsia
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan; (C.-H.H.); (H.-Y.S.)
- Department of Dietetics, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Hsiu-Yueh Su
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan; (C.-H.H.); (H.-Y.S.)
- Department of Dietetics, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Yi-Wen Chien
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 11031, Taiwan; (C.-H.H.); (H.-Y.S.)
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
14
|
Pham A, El-Kareh R, Myers F, Ohno-Machado L, Kuo TT. Predicting positive Clostridioides difficile test results using large-scale longitudinal data of demographics and medication history. Heliyon 2025; 11:e41350. [PMID: 39958729 PMCID: PMC11825254 DOI: 10.1016/j.heliyon.2024.e41350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 11/20/2024] [Accepted: 12/18/2024] [Indexed: 02/18/2025] Open
Abstract
Background Clostridioides difficile infection is a major health threat. Healthcare institutions have strong medical and financial incentives to keep infections under control. Blanket testing at admission is in general not recommended, and current predictive models either used moderate sample sizes, over-inflated the number of covariates, or chose non-interpretable algorithms. We aim to develop models using patient data to predict positive Clostridioides difficile test results with discrimination performance, interpretable results, and a reasonable number of covariates that reflect health over a long-time span. Materials and methods We processed records from 157,493 University of California San Diego Health patients seen between January 01, 2016-July 03, 2019 with at least 6 months of medication history, excluding pregnant women, patients under 18, and prisoners. Three models (Logistic Regression, Random Forest, and Ensemble) were constructed using hyper-parameters selected through 10-fold cross-validation. Model performance was measured by the Area Under the Receiver Operating Characteristic Curve (AUROC). The model coefficients' odds ratios and p-values were calculated for the Logistic Regression model, as were Gini indices for Random Forest. Decision boundary analysis was conducted using pair-wise false positive and false negative cases each model would predict at a specific threshold. Results Logistic Regression, Random Forest, and Ensemble models yielded test AUROCs of 0.839, 0.851, and 0.866, respectively. Significant covariates that may affect risk include age, immuno-compromised treatments, past antibiotic uses, and some medications for the gastrointestinal tract. Conclusions The models achieve high discrimination performance (AUROC >0.83). There is a general consensus among different analysis approaches regarding predictors that impact patients' chances of having a positive test, which may influence Clostridioides difficile risk, including features clinically proven to increase susceptibility. These human-interpretable models can help distinguish significant predictors that affect a patient's chance of testing positive, which may influence their Clostridioides difficile risk.
Collapse
Affiliation(s)
- Anh Pham
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Robert El-Kareh
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- UCSD Health System, San Diego, CA, USA
| | | | - Lucila Ohno-Machado
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA
| | - Tsung-Ting Kuo
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
Mullard RM, Sheedlo MJ. The N-terminus of the Clostridioides difficile transferase A component directs toxin activity and potency. mBio 2025; 16:e0240524. [PMID: 39611841 PMCID: PMC11708034 DOI: 10.1128/mbio.02405-24] [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: 08/06/2024] [Accepted: 11/12/2024] [Indexed: 11/30/2024] Open
Abstract
Clostridioides difficile infection is the leading cause of antibiotic-associated, hospital-acquired diarrhea in the USA; the pathology of which is mediated by toxins. The presence of a toxin known as the C. difficile Transferase (CDT) in some clinical isolates is linked to severe symptoms including increased incidence of reinfection and higher rates of mortality. Despite its apparent importance to C. difficile pathology, a mechanistic model of how CDT intoxicates cells remains incomplete. Here, we describe a motif composed of acidic and basic residues (the KDKEK motif) that is essential for toxin function. Using Cryogenic Electron Microscopy (Cryo-EM), we highlight an orientation of the KDKEK motif wherein the acidic residues engage structures thought to play an important role during toxin delivery. We thus present a model wherein these interactions prime CDT for entry into host cells. We expect that this model can be extrapolated to other bacterial toxins to understand how they enter cells.IMPORTANCEClostridioides difficile is the leading cause of hospital-acquired infectious diarrhea in the USA. The pathology that accompanies infection is triggered by toxins produced by the bacterium. One of these, the C. difficile Transferase (CDT), has been associated with poorer patient outcomes, although a direct connection to CDT activity has remained elusive. Herein, we present new insight into the mechanism of CDT intoxication and define two regions of the toxin as important for its activity. Moreover, we have generated mutants of CDT that retain the ability to assemble but can no longer intoxicate host cells. In the future, we expect these mutants will serve as valuable tools to help elucidate the role of CDT during infection.
Collapse
Affiliation(s)
- Robin M. Mullard
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Michael J. Sheedlo
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
16
|
Kullberg RFJ, Haak BW, Chanderraj R, Prescott HC, Dickson RP, Wiersinga WJ. Empirical antibiotic therapy for sepsis: save the anaerobic microbiota. THE LANCET. RESPIRATORY MEDICINE 2025; 13:92-100. [PMID: 39401510 DOI: 10.1016/s2213-2600(24)00257-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 01/07/2025]
Abstract
Antibiotics are fundamental in sepsis management; however, the optimal empirical treatment remains debated. Despite anaerobes rarely being the causative pathogen of sepsis, antibiotics targeting them are frequently used, which might lead to unintended consequences. Multiple studies have shown that depletion of commensal anaerobic gut microbes by anti-anaerobic antibiotics influences systemic immunity and is associated with increased mortality in patients with sepsis. However, this knowledge has not yet been translated into clinical practice. When considering empirical coverage of anaerobic pathogens in sepsis, most physicians advocate for a better-safe-than-sorry approach. In this Viewpoint, we argue that anti-anaerobic antibiotics could often result in being sorry rather than safe. We provide an overview of the limited necessity of anaerobic coverage and the potential detrimental effects of anaerobic depletion in sepsis. We aim to raise anaerobic awareness to reduce the unnecessary use of anti-anaerobic antibiotics in empirical sepsis treatment and improve patient outcomes.
Collapse
Affiliation(s)
- Robert F J Kullberg
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.
| | - Bastiaan W Haak
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Rishi Chanderraj
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Medicine Service, Infectious Diseases Section, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA; Weil Institute for Critical Care Research and Innovation, Ann Arbor, MI, USA
| | - Hallie C Prescott
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Veterans Affairs Center for Clinical Management Research, Ann Arbor, MI, USA
| | - Robert P Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA; Weil Institute for Critical Care Research and Innovation, Ann Arbor, MI, USA
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
17
|
Turello LA, Consul A, Yip C, Shen S, Seymour C, Geurink C, Alvarado I, Abel-Santos E. Differential gene expression analysis shows that cephalosporin resistance is intrinsic to Clostridioides difficile strain 630. J Antibiot (Tokyo) 2025; 78:113-125. [PMID: 39672901 DOI: 10.1038/s41429-024-00795-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/15/2024] [Accepted: 11/22/2024] [Indexed: 12/15/2024]
Abstract
Clostridioides difficile infection (CDI) is the most common nosocomial infection in the US. CDI has become a growing concern due to C. difficile's resistance to several antibiotics, including cephalosporins. Furthermore, patients administered cephalosporins are at higher risk of contracting CDI. Cephalosporins are β-lactam antibiotics, which prevent bacterial cell wall synthesis by inhibiting penicillin-binding proteins (PBPs). β-lactam-resistant bacteria evade these antibiotics by producing β-lactamases or by harboring low-affinity PBPs. A genomic analysis of C. difficile strain 630 identified 31 putative β-lactam resistance genes. Upon cefoxitin exposure, few C. difficile strain 630 putative antibiotic-resistant genes were overexpressed. Most notably, the β-lactamase blaCDD gene was upregulated approximately 600-fold, as previously reported. Deletion of the blaCDD locus did not change in cephalosporin susceptibility. Deletion of the second most upregulated gene, the PBP vanY, was also ineffective at decreasing cephalosporin resistance. Cefoxitin exposure of the C. difficile strain 630ΔblaCDD mutant did not increase upregulation of other putative antibiotic resistance genes compared to wildtype C. difficile strain 630. Transcriptomic analyses of wildtype C. difficile strain 630 exposed to cephradine, cefoxitin, ceftazidime, or cefepime revealed the shared upregulation of a putative heterodimeric ABC transporter encoded by loci CD630_04590 (ABC transporter ATP-binding protein) and CD630_04600 (ABC transporter permease). These genes are genomically located directly downstream of blaCDD (CD630_04580). The deletion mutant CD630_04600 remained resistant to a number of antibiotics. Thus, even though blaCDD, CD630_04590, and CD630_04600 are all upregulated when exposed to cephalosporins, they do not seem to be involved in antibiotic resistance in C. difficile strain 630.
Collapse
Affiliation(s)
- Lara A Turello
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Amber Consul
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Christopher Yip
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Shirley Shen
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, Las Vegas, NV, 89154-4004, USA
| | - Cale Seymour
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Corey Geurink
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, Las Vegas, NV, 89154-4004, USA
| | - Israel Alvarado
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA
- School of Medicine, Ponce Health Sciences University, Saint Louis, MO, 63103, USA
| | - Ernesto Abel-Santos
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, NV, USA.
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, Las Vegas, NV, 89154-4004, USA.
| |
Collapse
|
18
|
Schley K, Heinrich K, Moïsi JC, Häckl D, Obermüller D, Brestrich G, von Eiff C, Weinke T. Costs and Outcomes of Clostridioides difficile Infections in Germany: A Retrospective Health Claims Data Analysis. Infect Dis Ther 2025; 14:91-104. [PMID: 39565511 PMCID: PMC11782737 DOI: 10.1007/s40121-024-01075-1] [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: 07/22/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION Health claims data are a valuable resource for health services research, enabling analysis of the costs of hospitalizations, outpatient visits, procedures, and medications, and providing an improved understanding of the economic burden and underlying cost drivers for a given health condition. Since no recent data were available from Germany on the medical costs and clinical outcomes of Clostridioides difficile infections (CDI), this study assessed the economic burden of CDI and all-cause mortality in adults in Germany. METHODS A retrospective cohort study was conducted using a large, anonymized administrative health claims research database from Germany from which an age- and sex-representative sample of 4 million insured persons covered by approximately 60 statutory health insurances was extracted. Propensity score matching was conducted on age, sex, comorbidities, and antibiotic use to identify four matched controls (i.e., patients without CDI) for every eligible adult patient with CDI (i.e., case) in the study cohort. Costs, healthcare resource utilization, and CDI-attributable all-cause mortality were assessed. RESULTS Overall, there were 15,342 CDI cases in the study cohort. One-year mortality in CDI cases (45.7%) was more than fourfold that of matched non-CDI controls (11.0%). In the year following the index date, average mortality-adjusted medical costs per person-time for CDI cases were almost fivefold that of matched non-CDI controls, representing a cost difference of €31,459, mainly driven by inpatient treatment. Overall excess costs for CDI cases were estimated at approximately €1.6 billion within 1 year after diagnosis. CONCLUSIONS CDI in Germany is associated with a high clinical and economic burden, including significantly higher mortality, costs, and healthcare resource utilization, in patients with CDI versus their matched patients without CDI. This has important implications for patients, healthcare providers, and the healthcare system.
Collapse
Affiliation(s)
- Katharina Schley
- Pfizer Pharma GmbH, Friedrichstrasse 110, 10117, Berlin, Germany.
| | - Kirstin Heinrich
- Pfizer Inc., Patient and Health Impact, New York, NY, 10017, USA
| | - Jennifer C Moïsi
- Pfizer Vaccines, Medical Development and Scientific/Clinical Affairs, 23 Av du Dr Lannelongue, 75014, Paris, France
| | | | - Dominik Obermüller
- InGef-Institute for Applied Health Research Berlin GmbH, Otto-Ostrowski-Str. 5, 10249, Berlin, Germany
| | - Gordon Brestrich
- Pfizer Pharma GmbH, Friedrichstrasse 110, 10117, Berlin, Germany
| | | | - Thomas Weinke
- Ernst Von Bergmann Klinikum, Charlottenstraße 72, 14467, Potsdam, Germany
| |
Collapse
|
19
|
Hsia CH, Su HY, Chen YH, Chuang HC, Chien YW. Effects of probiotics in elderly hospitalized tube-fed patients with antibiotics use. BMC Gastroenterol 2024; 24:467. [PMID: 39702125 DOI: 10.1186/s12876-024-03561-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Several studies revealed the beneficial effects of probiotics against the incidence of antibiotic-associated diarrhea of hospitalized patients but it is rarely to assess the nutrition status. This study investigated the effects of probiotics in elderly hospitalized tube-fed patients with antibiotics use and is the first study that concerns the nutritional status among these patients. METHODS Elderly hospitalized tube-fed patients who were using antibiotics were recruited. Probiotics were given within 48 h after their first antibiotic therapy, and then twice daily 2 h after consuming antibiotics and a meal; the probiotics were continued to use for an additional 7 days after completion of antibiotics therapy. Anthropometric data, laboratory data, medication records, nutritional status, nutrition intake and data on stool form were collected. RESULTS Twenty-nine patients served as probiotic group. 11 patients completed the study in both groups. In probiotic group, the stool form was found to exhibit no significant differences between the beginning and end of antibiotics therapy (5.5 ± 0.8 vs 5.1 ± 1.1, p = 0.21), but the stool frequency significantly decreased (2.0 ± 1.0 vs 1.6 ± 0.7, p = 0.05). In control group, the stool form between the beginning and end of antibiotics therapy exhibited significant improvement (5.6 ± 1.4 vs 4.5 ± 1.4, p = 0.01), but not in the frequency (2.7 ± 2.1 vs 2.4 ± 1.5, p = 0.1). The initial NRS 2002 score of the probiotic and control groups were similar. (3.6 ± 1.7 vs 3.7 ± 1.8, p = 1.00), and their nutrition status both significantly improved during the last visit before discharged (2.6 ± 0.9 vs 2.9 ± 1.3). CONCLUSION Probiotic supplementation in elderly hospitalized tube-fed patients significantly reduced stool frequency during antibiotic treatment. Improvements in stool form were observed only during the follow-up period. Nutritional status remained stable, with patients' nutritional needs adequately met throughout the study.
Collapse
Affiliation(s)
- Chu-Hsuan Hsia
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, 11031, Taiwan, ROC
- Department of Dietetics, Taipei Medical University Hospital, Taipei, 11031, Taiwan, ROC
- Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan, ROC
| | - Hsiu-Yueh Su
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, 11031, Taiwan, ROC
- Department of Dietetics, Taipei Medical University Hospital, Taipei, 11031, Taiwan, ROC
- Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan, ROC
| | - Yue-Hwa Chen
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, 11031, Taiwan, ROC
- School of Food Safety, Taipei Medical University, Taipei, 11031, Taiwan, ROC
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Research Center of Food Safety Inspection and Function Development, Taipei Medical University, Taipei, 11031, Taiwan, ROC
| | - Han-Chuan Chuang
- Department of Infectious Disease, Taipei Medical University Hospital, Taipei, 11031, Taiwan, ROC
| | - Yi-Wen Chien
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
- Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan, ROC.
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
| |
Collapse
|
20
|
Donskey CJ, Dubberke ER, Klein NP, Liles EG, Szymkowiak K, Wilcox MH, Lawrence J, Bouguermouh S, Zhang H, Koury K, Bailey R, Smith HM, Lockhart S, Lamberth E, Kalina WV, Pride MW, Webber C, Anderson AS, Jansen KU, Gruber WC, Kitchin N. CLOVER (CLOstridium difficile Vaccine Efficacy tRial) Study: A Phase 3, Randomized Trial Investigating the Efficacy and Safety of a Detoxified Toxin A/B Vaccine in Adults 50 Years and Older at Increased Risk of Clostridioides difficile Infection. Clin Infect Dis 2024; 79:1503-1511. [PMID: 39180325 PMCID: PMC11650871 DOI: 10.1093/cid/ciae410] [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: 12/08/2023] [Revised: 05/20/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Clostridioides difficile infection (CDI) causes substantial mortality and healthcare burden. We assessed the detoxified toxin-A/B PF-06425090 vaccine for primary CDI prevention. METHODS This phase 3 observer-blinded study randomized (1:1) ≥50-year-olds at increased CDI risk (N = 17 535) to receive 3 PF-06425090 or placebo doses (0, 1, and 6 months). Primary end points were first CDI episode (≥3 unformed stools within 24 hours; central laboratory-confirmed toxin A/B positive) ≥14 days post-dose 3 (PD3; first primary) and post-dose 2 (PD2; second primary). CDI duration, need for CDI-related medical attention (secondary end points), and antibiotic use (post hoc analysis) PD3 were evaluated. Tolerability and safety were assessed. RESULTS The primary end point was not met (17 PF-06425090 and 25 placebo recipients had first CDI episode ≥14 days PD3 [vaccine efficacy (VE) = 31.0% (96.4% confidence interval [CI], -38.7% to 66.6%)]; 24 PF-06425090 and 34 placebo recipients had first CDI episode ≥14 days PD2 [VE = 28.6% (96.4% CI, -28.4% to 61.0%)]. Median CDI duration was lower with PF-06425090 (1 day) versus placebo (4 days; 2-sided nominal P = .02). Of participants with first CDI episode, 0 PF-06425090 and 11 placebo recipients sought CDI-related medical attention (post hoc analysis estimated VE = 100%; 95% CI, 59.6% to 100.0%) and 0 PF-06425090 and 10 placebo recipients required antibiotic treatment (VE = 100%; 95% CI, 54.8% to 100.0%). Local reactions were more frequent in PF-06425090 recipients, and systemic events were generally similar between groups; most were mild to moderate. Adverse event rates were similar between groups. CONCLUSIONS Three PF-06425090 doses were safe and well tolerated. Although the primary end point was not met, PF-06425090 reduced symptom duration, CDI that required medical attention, and CDI-directed antibiotic treatment, highlighting its potential to reduce CDI-associated healthcare burden. CLINICAL TRIALS REGISTRATION NCT03090191.
Collapse
Affiliation(s)
- Curtis J Donskey
- Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Erik R Dubberke
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Nicola P Klein
- Kaiser Permanente Vaccine Study Center, Oakland, California, USA
| | | | | | - Mark H Wilcox
- Leeds Teaching Hospitals NHS Trust and Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Stolz BJ, Abouelkhair AA, Seleem MN. Screening novel antiviral compounds to treat Clostridioides difficile infections. PLoS One 2024; 19:e0309624. [PMID: 39671442 PMCID: PMC11642915 DOI: 10.1371/journal.pone.0309624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/27/2024] [Indexed: 12/15/2024] Open
Abstract
Clostridioides difficile is a major cause of nosocomial infections, often associated with individuals who have gut dysbiosis from previous antibiotic therapies. C. difficile infections (CDI) have a high recurrence rate and impose significant financial and mortality burdens on the healthcare system. Therefore, novel anti-C. difficile drugs are urgently needed to treat and reduce the severity and recurrence of infection. In this study, we screened a library of 618 antiviral drugs to identify a potential candidate for repurposing as novel anti-C. difficile therapeutics. Following our preliminary screening, we identified 9 novel compounds that inhibited C. difficile at a concentration of 16 μM or lower. Among these, 4 antiviral compounds demonstrated the most potent anti-C. difficile activity against a panel of 15 C. difficile isolates, with minimum inhibitory concentrations (MICs) comparable to the drug of choice, vancomycin. These include rottlerin (MIC50 = 0.25 μg/mL), α-mangostin (MIC50 = 1 μg/mL), dryocrassin ABBA (MIC50 = 1 μg/mL), and obefazimod (MIC50 = 4 μg/mL). All exhibited minimal to no activity against representative members of the human gut microbiota. Interestingly, α-mangostin, a natural xanthone derived from the mangosteen fruit, exhibited strong bactericidal action, clearing a high inoculum of C. difficile in less than an hour. All other drugs exhibited bacteriostatic activity. Given their characteristics, these compounds show great promise as novel treatments for CDI.
Collapse
Affiliation(s)
- Brice J. Stolz
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Ahmed A. Abouelkhair
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Mohamed N. Seleem
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| |
Collapse
|
22
|
Wu LH, Wang JL, Liu YH, Su CC, Yang YHK, Lin SJ, Cheng CL. Hospitalized patients on proton pump inhibitors for stress ulcer prophylaxis have a higher risk of Clostridioides difficile infection compared with those on histamine-2 receptor antagonists. J Hosp Infect 2024; 154:9-17. [PMID: 39369994 DOI: 10.1016/j.jhin.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Previous studies on Clostridioides difficile infection (CDI) in proton pump inhibitor (PPI) users generally enrolled a heterogeneous population and did not include a control group of histamine H2 receptor antagonists (H2RAs) users or adjust for confounding variables, such as previous antibiotics. It is uncertain whether hospitalized patients using PPIs for stress ulcer prophylaxis (SUP) are at a higher risk of CDI compared with those using H2RAs. This study aimed to compare the association between CDI and the usage of antisecretory drugs (ASDs): PPIs and H2RAs, for SUP among hospitalized patients, and the impact of the duration of their use on CDI. METHODS In this nationwide population-based cohort study using the Taiwan National Health Insurance Database, hospitalized patients using ASDs for SUP were identified between 2017 and 2018. A total of 63,266 and 69,269 individuals were included in the PPI and H2RA groups, respectively. The primary endpoint was a 90-day monitoring of CDI occurrence. FINDINGS The incidences of CDI were 1.6/10,000 and 0.5/10,000 person-days in the PPIs and H2RAs groups, respectively. After adjusting for confounding factors, the risk of infection in the PPIs group remained significantly higher than in the H2RAs group (hazard ratio (HR), 2.49; 95% confidence interval (CI), 1.63-3.81). In the subgroup analysis, during hospitalization, the risk of CDI for patients using high-risk antibiotics or admitted to the intensive care unit (ICU), as well as patients with immunodeficiency, using PPIs for SUP, was higher than using H2RAs. Furthermore, the risk of CDI was higher in patients using ASDs for durations >14 days than in those using them for <7 days (adjusted HR, 3.66; 95% CI, 2.34-5.75). CONCLUSIONS The risk of occurrence CDI for hospitalized patients using PPIs for SUP was higher than using H2RAs. It is recommended not to exceed 14 days of any gastric ASDs for SUP during hospitalization, especially for patients who have used high-risk antibiotics, have been admitted to the ICU, or have immunodeficiency.
Collapse
Affiliation(s)
- L-H Wu
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan City, Taiwan; Department of Pharmacy, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - J-L Wang
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan City, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Y-H Liu
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan City, Taiwan
| | - C-C Su
- Clinical Innovation and Research Center, National Cheng Kung University Hospital, Tainan City, Taiwan
| | - Y-H K Yang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan City, Taiwan
| | - S-J Lin
- Department of Pharmacy Systems, Outcomes and Policy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - C-L Cheng
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan City, Taiwan; Department of Pharmacy, National Cheng Kung University Hospital, Tainan City, Taiwan.
| |
Collapse
|
23
|
Karp J, Edman-Wallér J, Jacobsson G. Duration from start of antibiotic exposure to onset of Clostridioides difficile infection for different antibiotics in a non-outbreak setting. Infect Dis (Lond) 2024; 56:1049-1056. [PMID: 39023136 DOI: 10.1080/23744235.2024.2375602] [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: 04/17/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Antibiotic treatment is a well-known risk factor for Clostridioides difficile infection (CDI). The time from start of antibiotic exposure to onset of CDI for different antibiotics is sparsely studied. CDI with onset in the community is often treatable without in-hospital care while CDI patients treated in hospital need isolation, resulting in higher costs and infection control measures. OBJECTIVES To determine the time from start of antibiotic exposure to onset of healthcare facility-associated CDI for different antibiotics. METHODS Time between antibiotic exposure and disease onset was evaluated retrospectively with chart reading in a two-centre Swedish setting. A case was attributed to an antibiotic group if this represented more than 2/3 of total antibiotic exposure 30 days before onset of CDI. RESULTS Cephalosporins caused CDI faster (mean 7.6 days), and more often during ongoing antibiotic therapy (81% of the cases) than any other antibiotic group. All other common agents had between 2-3 times longer period between start of exposure to onset of CDI (quinolones more than 3 times). CONCLUSIONS The time gap between antibiotic exposure and onset of CDI is markedly different between different antibiotics. Decreased cephalosporin use could delay onset of healthcare facility-associated CDI and limit infections with onset within the hospital. This might decrease costs for inpatient care, need of infection control measures and shortage of beds in the hospital.
Collapse
Affiliation(s)
- Johan Karp
- Department of Infectious Diseases, Skaraborg Hospital, Skövde, Sweden
- Centre for Antibiotic Resistance Research (CARe), Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Skaraborgsinstitutet, Skövde, Sweden
| | - Jon Edman-Wallér
- Centre for Antibiotic Resistance Research (CARe), Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Infection Prevention and Control, Södra Älvsborg Hospital, Borås, Sweden
| | - Gunnar Jacobsson
- Department of Infectious Diseases, Skaraborg Hospital, Skövde, Sweden
- Centre for Antibiotic Resistance Research (CARe), Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
24
|
Tan C, xiao Y, Liu T, Chen S, Zhou J, Zhang S, Hu Y, Wu A, Li C. Development of multi-epitope mRNA vaccine against Clostridioides difficile using reverse vaccinology and immunoinformatics approaches. Synth Syst Biotechnol 2024; 9:667-683. [PMID: 38817826 PMCID: PMC11137598 DOI: 10.1016/j.synbio.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/28/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
Clostridioides difficile (C. difficile), as the major pathogen of diarrhea in healthcare settings, has become increasingly prevalent within community populations, resulting in significant morbidity and mortality. However, the therapeutic options for Clostridioides difficile infection (CDI) remain limited, and as of now, no authorized vaccine is available to combat this disease. Therefore, the development of a novel vaccine against C. difficile is of paramount importance. In our study, the complete proteome sequences of 118 strains of C. difficile were downloaded and analyzed. We found four antigenic proteins that were highly conserved and can be used for epitope identification. We designed two vaccines, WLcd1 and WLcd2, that contain the ideal T-cell and B-cell epitopes, adjuvants, and the pan HLA DR-binding epitope (PADRE) sequences. The biophysical and chemical assessments of these vaccine candidates indicated that they were suitable for immunogenic applications. Molecular docking analyses revealed that WLcd1 bonded with higher affinity to Toll-like receptors (TLRs) than WLcd2. Furthermore, molecular dynamics (MD) simulations, performed using Gmx_MMPBSA v1.56, confirmed the binding stability of WLcd1 with TLR2 and TLR4. The preliminary findings suggested that this multi-epitope vaccine could be a promising candidate for protection against CDI; however, experimental studies are necessary to confirm these predictions.
Collapse
Affiliation(s)
- Caixia Tan
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Yuanyuan xiao
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Ting Liu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Siyao Chen
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Juan Zhou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Sisi Zhang
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Yiran Hu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Anhua Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| | - Chunhui Li
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
- National Clinical Research Center for Geriatric Disorders (XiangYa Hospital), Changsha, Hunan Province, 410008, China
| |
Collapse
|
25
|
Siroglavic M, Higgins PG, Kanizaj L, Ferencak I, Juric D, Augustin G, Budimir A. Whole-Genome Sequencing-Based Characterization of Clostridioides difficile Infection Cases at the University Hospital Centre Zagreb. Microorganisms 2024; 12:2434. [PMID: 39770637 PMCID: PMC11676685 DOI: 10.3390/microorganisms12122434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025] Open
Abstract
We investigated the intra-hospital distribution of C. difficile strains by whole-genome sequencing (WGS) of isolates collected in 2022 at the University Hospital Centre (UHC) Zagreb. In total, 103 patients with first-episode CDI in 2022 at UHC Zagreb were included, based on the screening stool antigen test for GDH (RidaQuick CD GDH; R-Biopharm AG, Germany), confirmed by Eazyplex C. difficile assays (Eazyplex CD assay; AmplexDiagnostics GmbH, Germany) specific for A, B, and binary toxins. Demographic and clinical data were retrospectively analyzed from electronic medical records. All samples were subjected to WGS analysis. Genetic clusters were formed from isolates with no more than six allelic differences according to core genome MLST. We identified six clusters containing 2-59 isolates with 15 singletons and 30 instances of possible intra-hospital transmission, mostly in the COVID-19 ward. WGS analysis proved useful in identifying clusters of isolates connecting various patient wards with possible transmission routes in the hospital setting. It could be used to support local and national surveillance of CDI infections and their transmission pathways.
Collapse
Affiliation(s)
- Marko Siroglavic
- Department of Clinical Microbiology, Infection Prevention and Control, University Hospital Centre Zagreb, Kispaticeva st. 12, 10000 Zagreb, Croatia; (M.S.); (L.K.); (A.B.)
| | - Paul G. Higgins
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Goldenfelsstraße 19-21, 50935 Cologne, Germany;
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, 50935 Cologne, Germany
| | - Lucija Kanizaj
- Department of Clinical Microbiology, Infection Prevention and Control, University Hospital Centre Zagreb, Kispaticeva st. 12, 10000 Zagreb, Croatia; (M.S.); (L.K.); (A.B.)
| | - Ivana Ferencak
- Department of Microbiology, Croatian Institute of Public Health, Rockefeller st. 7, 10000 Zagreb, Croatia; (I.F.); (D.J.)
| | - Dragan Juric
- Department of Microbiology, Croatian Institute of Public Health, Rockefeller st. 7, 10000 Zagreb, Croatia; (I.F.); (D.J.)
| | - Goran Augustin
- Department of Surgery, University Hospital Centre Zagreb, Kispaticeva st. 12, 10000 Zagreb, Croatia
| | - Ana Budimir
- Department of Clinical Microbiology, Infection Prevention and Control, University Hospital Centre Zagreb, Kispaticeva st. 12, 10000 Zagreb, Croatia; (M.S.); (L.K.); (A.B.)
- Department of Microbiology and Parasitology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
26
|
Franza M, Varricchio R, Alloisio G, De Simone G, Di Bella S, Ascenzi P, di Masi A. Zebrafish ( Danio rerio) as a Model System to Investigate the Role of the Innate Immune Response in Human Infectious Diseases. Int J Mol Sci 2024; 25:12008. [PMID: 39596075 PMCID: PMC11593600 DOI: 10.3390/ijms252212008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
The zebrafish (Danio rerio) has emerged as a valuable model for studying host-pathogen interactions due to its unique combination of characteristics. These include extensive sequence and functional conservation with the human genome, optical transparency in larvae that allows for high-resolution visualization of host cell-microbe interactions, a fully sequenced and annotated genome, advanced forward and reverse genetic tools, and suitability for chemical screening studies. Despite anatomical differences with humans, the zebrafish model has proven instrumental in investigating immune responses and human infectious diseases. Notably, zebrafish larvae rely exclusively on innate immune responses during the early stages of development, as the adaptive immune system becomes fully functional only after 4-6 weeks post-fertilization. This window provides a unique opportunity to isolate and examine infection and inflammation mechanisms driven by the innate immune response without the confounding effects of adaptive immunity. In this review, we highlight the strengths and limitations of using zebrafish as a powerful vertebrate model to study innate immune responses in infectious diseases. We will particularly focus on host-pathogen interactions in human infections caused by various bacteria (Clostridioides difficile, Staphylococcus aureus, and Pseudomonas aeruginosa), viruses (herpes simplex virus 1, SARS-CoV-2), and fungi (Aspergillus fumigatus and Candida albicans).
Collapse
Affiliation(s)
- Maria Franza
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Romualdo Varricchio
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Giulia Alloisio
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Giovanna De Simone
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy;
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
- Accademia Nazionale dei Lincei, 00165 Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
- Centro Linceo Interdisciplinare “Beniamino Segre”, Accademia Nazionale dei Lincei, 00165 Roma, Italy
| |
Collapse
|
27
|
Mancinetti F, Marinelli A, Boccardi V, Mecocci P. Challenges of infectious diseases in older adults: From immunosenescence and inflammaging through antibiotic resistance to management strategies. Mech Ageing Dev 2024; 222:111998. [PMID: 39447983 DOI: 10.1016/j.mad.2024.111998] [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: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Infectious diseases in older adults present a significant challenge to the healthcare system, marked by increased morbidity, mortality, and rising costs of care. Age-related changes (ARCs) in the immune system, including immunosenescence and inflammaging, contribute to heightened susceptibility to severe infections and reduced vaccine responsiveness. Additionally, alterations in the normal microbial flora due to aging and factors such as antibiotic therapy predispose older individuals to infections and age-related diseases. Changes in body composition also affect the pharmacokinetics and pharmacodynamics of drugs, complicating the management of antibiotics and leading to potential overdoses, adverse drug reactions, or underdoses that foster antibiotic resistance. The inappropriate use of antibiotics has exacerbated the emergence of multidrug-resistant pathogens, posing a critical global concern. This narrative review provides an overview of immunosenescence and inflammaging and focuses on three major infectious diseases affecting older adults: bacterial pneumonia, urinary tract infections, and Clostridium difficile infections. Through this exploration, we aim to highlight the need for targeted approaches in managing infectious diseases in the aging population.
Collapse
Affiliation(s)
- Francesca Mancinetti
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia-Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Anna Marinelli
- Clinical of Internal Medicine, Department of Medical Surgical and Health Science, University of Trieste, Cattinara Hospital, Trieste, Italy
| | - Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia-Santa Maria della Misericordia Hospital, Perugia, Italy.
| | - Patrizia Mecocci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia-Santa Maria della Misericordia Hospital, Perugia, Italy; Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
De-la-Rosa-Martinez D, Vilar-Compte D, Martínez-Rivera N, Ochoa-Hein E, Morfin-Otero R, Rangel-Ramírez ME, Garciadiego-Fossas P, Mosqueda-Gómez JL, Rodríguez Zulueta AP, Medina-Piñón I, Franco-Cendejas R, Alfaro-Rivera CG, Rivera-Martínez NE, Mendoza-Barragán J, López-Romo AE, Manríquez-Reyes M, Martínez-Oliva DH, Flores-Treviño S, Azamar-Marquez JM, Valverde-Ramos LN, Nieto-Saucedo JR, Aguirre-Díaz SA, Camacho-Ortiz A. Multicenter study on Clostridioides difficile infections in Mexico: exploring the landscape. Infect Control Hosp Epidemiol 2024:1-8. [PMID: 39431358 DOI: 10.1017/ice.2024.153] [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 This study aims to outline Clostridioides difficile infection (CDI) trends and outcomes in Mexican healthcare facilities during the COVID-19 pandemic. DESIGN Observational study of case series. SETTING Sixteen public hospitals and private academic healthcare institutions across eight states in Mexico from January 2016 to December 2022. PATIENTS CDI patients. METHODS Demographic, clinical, and laboratory data of CDI patients were obtained from clinical records. Cases were classified as community or healthcare-associated infections, with incidence rates calculated as cases per 10,000 patient days. Risk factors for 30-day all-cause mortality were analyzed by multivariate logistic regression. RESULTS We identified 2,356 CDI cases: 2,118 (90%) were healthcare-associated, and 232 (10%) were community-associated. Common comorbidities included hypertension, diabetes, and cancer. Previous high use of proton-pump inhibitors, steroids, and antibiotics was observed. Recurrent infection occurred in 112 (5%) patients, and 30-day mortality in 371 (16%). Risk factors associated with death were a high Charlson score, prior use of steroids, concomitant use of antibiotics, leukopenia, leukocytosis, elevated serum creatine, hypoalbuminemia, septic shock or abdominal sepsis, and SARS-CoV-2 coinfection. The healthcare-associated CDI incidence remained stable at 4.78 cases per 10,000 patient days during the pre-and pandemic periods. However, the incidence was higher in public hospitals. CONCLUSIONS Our study underscores the need for routine epidemiology surveillance and standardized CDI classification protocols in Mexican institutions. Though CDI rates in our country align with those in some European countries, disparities between public and private healthcare sectors emphasize the importance of targeted interventions.
Collapse
Affiliation(s)
- Daniel De-la-Rosa-Martinez
- Instituto Nacional de Cancerología, Mexico City, Mexico
- F. I. Proctor Foundation, University of California San Francisco, San Francisco, USA
| | | | | | - Eric Ochoa-Hein
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rayo Morfin-Otero
- Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | | | | | - Juan Luis Mosqueda-Gómez
- Hospital Regional de Alta Especialidad del Bajío, Servicios de Salud del Instituto Mexicano de Seguro Social para el Bienestar (IMSS-BIENESTAR), Guanajuato, Mexico
| | | | | | | | | | - Norma Eréndira Rivera-Martínez
- Hospital Regional de Alta Especialidad de Oaxaca, Servicios de Salud del Instituto Mexicano de Seguro Social para el Bienestar (IMSS-BIENESTAR), Oaxaca, Mexico
| | | | | | | | | | | | | | | | - José Raúl Nieto-Saucedo
- Hospital Regional de Alta Especialidad del Bajío, Servicios de Salud del Instituto Mexicano de Seguro Social para el Bienestar (IMSS-BIENESTAR), Guanajuato, Mexico
| | | | | |
Collapse
|
29
|
Lee A, Yoo JS, Yoon EJ. Gut Microbiota and New Microbiome-Targeted Drugs for Clostridioides difficile Infections. Antibiotics (Basel) 2024; 13:995. [PMID: 39452261 PMCID: PMC11505460 DOI: 10.3390/antibiotics13100995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Clostridioides difficile is a major causative pathogen for antibiotic-associated diarrhea and C. difficile infections (CDIs) may lead to life-threatening diseases in clinical settings. Most of the risk factors for the incidence of CDIs, i.e., antibiotic use, treatment by proton pump inhibitors, old age, and hospitalization, are associated with dysbiosis of gut microbiota and associated metabolites and, consequently, treatment options for CDIs include normalizing the composition of the intestinal microbiome. In this review, with an introduction to the CDI and its global epidemiology, CDI-associated traits of the gut microbiome and its metabolites were reviewed, and microbiome-targeting treatment options were introduced, which was approved recently as a new drug by the United States Food and Drug Administration (U.S. FDA), rather than a medical practice.
Collapse
Affiliation(s)
| | | | - Eun-Jeong Yoon
- Division of Antimicrobial Resistance Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju-si 28159, Republic of Korea
| |
Collapse
|
30
|
Jacob R, Chu V, Ng W, Williams AJ, Connor S. Treatment outcomes of mild to moderate Clostridioides difficile infection in inflammatory bowel disease: an Australian experience. Intern Med J 2024. [PMID: 39422325 DOI: 10.1111/imj.16545] [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: 04/28/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The incidence of Clostridioides difficile infection (CDI) in inflammatory bowel disease (IBD) is rising and associated with adverse outcomes. Optimal treatment for CDI in IBD remains unknown, with various organisations suggesting vancomycin as first-line therapy. AIM To compare treatment outcomes for metronidazole versus vancomycin in mild to moderate CDI in IBD. METHODS A retrospective analysis of IBD patients with CDI between January 2015 and December 2019 was conducted. Laboratory data and clinical outcomes were examined. RESULTS Forty-seven discrete episodes of CDI in 34 patients occurred during the study period. Fifty-three per cent (18) had Crohn's disease, 44% (15) ulcerative colitis and 3% (1) IBD unclassified. Six per cent (3/47) of CDI cases were severe and excluded. In 68% (30/44) of mild to moderate CDI, metronidazole was prescribed, with treatment failure in 20% (6/30), CDI recurrence in 13% (4/30) and 20% (6/30) experiencing a further CDI episode. Comparatively, vancomycin was prescribed in 23% (10/44) without any treatment failures; however, 10% (1/10) had CDI recurrence and 40% (4/10) experienced another CDI episode. No statistically significant difference was noted between metronidazole and vancomycin therapy for treatment failure (P = 0.31), CDI recurrence (P = 1.0) or further CDI episodes (P = 0.23). Proton-pump inhibitor therapy was the only risk factor associated with a higher rate of the composite outcome and remained significant on multivariate logistic regression (odds ratio 12.99 (95% confidence interval = 1.21-139.97), P = 0.03). CONCLUSION Metronidazole compared to vancomycin for treatment of mild to moderate CDI in IBD is effective however may be associated with higher rates of treatment failure.
Collapse
Affiliation(s)
- Rachael Jacob
- Department of Gastroenterology & Hepatology, Liverpool Hospital, Sydney, New South Wales, Australia
| | | | - Watson Ng
- Department of Gastroenterology & Hepatology, Liverpool Hospital, Sydney, New South Wales, Australia
| | - Astrid-Jane Williams
- Department of Gastroenterology & Hepatology, Liverpool Hospital, Sydney, New South Wales, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | - Susan Connor
- Department of Gastroenterology & Hepatology, Liverpool Hospital, Sydney, New South Wales, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- The Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
31
|
Marreddy RKR, Phelps GA, Churion K, Picker J, Powell R, Cherian PT, Bowling JJ, Stephan CC, Lee RE, Hurdle JG. Chemical genetic analysis of enoxolone inhibition of Clostridioides difficile toxin production reveals adenine deaminase and ATP synthase as antivirulence targets. J Biol Chem 2024; 300:107839. [PMID: 39343002 PMCID: PMC11566853 DOI: 10.1016/j.jbc.2024.107839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Toxins TcdA and TcdB are the main virulence factors of Clostridioides difficile, a leading cause of hospital-acquired diarrhea. Despite their importance, there is a significant knowledge gap of druggable targets for inhibiting toxin production. To address this, we screened nonantibiotic phytochemicals to identify potential chemical genetic probes to discover antivirulence drug targets. This led to the identification of 18β-glycyrrhetinic acid (enoxolone), a licorice metabolite, as an inhibitor of TcdA and TcdB biosynthesis. Using affinity-based proteomics, potential targets were identified as ATP synthase subunit alpha (AtpA) and adenine deaminase (Ade, which catalyzes conversion of adenine to hypoxanthine in the purine salvage pathway). To validate these targets, a multifaceted approach was adopted. Gene silencing of ade and atpA inhibited toxin biosynthesis, while surface plasmon resonance and isothermal titration calorimetry molecular interaction analyses revealed direct binding of enoxolone to Ade. Metabolomics demonstrated enoxolone induced the accumulation of adenosine, while depleting hypoxanthine and ATP in C. difficile. Transcriptomics further revealed enoxolone dysregulated phosphate uptake genes, which correlated with reduced cellular phosphate levels. These findings suggest that enoxolone's cellular action is multitargeted. Accordingly, supplementation with both hypoxanthine and triethyl phosphate, a phosphate source, was required to fully restore toxin production in the presence of enoxolone. In conclusion, through the characterization of enoxolone, we identified promising antivirulence targets that interfere with nucleotide salvage and ATP synthesis, which may also block toxin biosynthesis.
Collapse
Affiliation(s)
- Ravi K R Marreddy
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Gregory A Phelps
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Kelly Churion
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Jonathan Picker
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Reid Powell
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Philip T Cherian
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - John J Bowling
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Clifford C Stephan
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Julian G Hurdle
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, USA.
| |
Collapse
|
32
|
Sauvat L, Verhoeven PO, Gagnaire J, Berthelot P, Paul S, Botelho-Nevers E, Gagneux-Brunon A. Vaccines and monoclonal antibodies to prevent healthcare-associated bacterial infections. Clin Microbiol Rev 2024; 37:e0016022. [PMID: 39120140 PMCID: PMC11391692 DOI: 10.1128/cmr.00160-22] [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: 08/10/2024] Open
Abstract
SUMMARYHealthcare-associated infections (HAIs) represent a burden for public health with a high prevalence and high death rates associated with them. Pathogens with a high potential for antimicrobial resistance, such as ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) and Clostridioides difficile, are responsible for most HAIs. Despite the implementation of infection prevention and control intervention, globally, HAIs prevalence is stable and they are mainly due to endogenous pathogens. It is undeniable that complementary to infection prevention and control measures, prophylactic approaches by active or passive immunization are needed. Specific groups at-risk (elderly people, chronic condition as immunocompromised) and also healthcare workers are key targets. Medical procedures and specific interventions are known to be at risk of HAIs, in addition to hospital environmental exposure. Vaccines or monoclonal antibodies can be seen as attractive preventive approaches for HAIs. In this review, we present an overview of the vaccines and monoclonal antibodies in clinical development for prevention of the major bacterial HAIs pathogens. Based on the current state of knowledge, we look at the challenges and future perspectives to improve prevention by these means.
Collapse
Affiliation(s)
- Léo Sauvat
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Paul O Verhoeven
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Agents and Hygiene, University-Hospital of Saint-Etienne, Saint-Etienne, France
| | - Julie Gagnaire
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Philippe Berthelot
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Amandine Gagneux-Brunon
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
33
|
Qin P, Moore MJ, Jung S, Fukazawa T, Yamasaki N, Chatterjee S, Wu Z, Boger DL. Tetrachloromaxamycins: Divergent Total Synthesis and Initial Assessments. J Org Chem 2024; 89:12701-12710. [PMID: 39169612 PMCID: PMC11380578 DOI: 10.1021/acs.joc.4c01927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Divergent total syntheses of binding pocket and peripherally modified tetrachlorovancomycins, a non-native synthetic glycopeptide, and their evaluation are disclosed. Central to the approach is the synthesis of a single late-stage intermediate that bears a residue 4 thioamide ([Ψ[C(═S)NH]Tpg4]tetrachlorovancomycin (3), LLS 15 steps, 14% overall) as a precursor to either of two key pocket modifications and their pairing with any combination of two peripheral modifications conducted without protecting groups. A stereochemical simplification achieved by the addition of two aryl chlorides removes two synthetically challenging atropisomer centers in native glycopeptides and streamlines the synthesis. Key features include in a convergent epimerization-free thioacylation of the AB ring system amine with an N-thioacylbenzotriazolyl DE tetrapeptide (85%) followed by simultaneous room-temperature SNAr macrocyclizations of the CD and DE ring systems (96%). The approach provided 3 from which [Ψ[C(═N)NH]Tpg4]tetrachlorovancomycin (4) and [Ψ(CH2NH)Tpg4]tetrachlorovancomycin (5) were prepared in a single-step and bear binding pocket modifications that convey dual d-Ala-d-Ala/d-Lac ligand binding to overcome vancomycin resistance. The newest maxamycin members are disclosed, bearing two additional peripheral modifications that introduce two independent synergistic MOAs that do not rely on native ligand binding for activity. Ligand binding properties of pocket-modified tetrachlorovancomycins 3-5, antibacterial activity of a key compound series, and PK assessments of two tetrachloromaxamycins are reported.
Collapse
Affiliation(s)
| | | | - Sunna Jung
- Departments of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N., Torrey Pines Road, La Jolla, CA 92037, USA
| | - Takumi Fukazawa
- Departments of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N., Torrey Pines Road, La Jolla, CA 92037, USA
| | - Naoto Yamasaki
- Departments of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N., Torrey Pines Road, La Jolla, CA 92037, USA
| | - Shreyosree Chatterjee
- Departments of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N., Torrey Pines Road, La Jolla, CA 92037, USA
| | - Zhi–Chen Wu
- Departments of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N., Torrey Pines Road, La Jolla, CA 92037, USA
| | - Dale L. Boger
- Departments of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N., Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
34
|
Nagesh VK, Tran HHV, Elias D, Kianifar Aguilar I, Sethi T, Menon A, Mansour C, Furman F, Tsotsos K, Subar T, Auda A, Sidiqui A, Lamar J, Wadhwani N, Dey S, Lo A, Atoot A, Weissman S, Sifuentes H, Bangolo AI. Therapeutics involved in managing initial and recurrent Clostridium difficile infection: An updated literature review. World J Gastrointest Pharmacol Ther 2024; 15:95467. [PMID: 39281262 PMCID: PMC11401021 DOI: 10.4292/wjgpt.v15.i5.95467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Clostridium difficile infection (CDI) has been increasing due to the effect of recurrent hospitalizations. The use of antibiotics has been shown to alter the gut microbiome and lead to CDIs. The treatment is limited to three major antibiotics; however, the incidence of recurrent CDIs has been increasing and drug resistance is a major concern. This aspect is a growing concern in modern medicine especially in the elderly population, critical care patients, and immunocompromised individuals who are at high risk of developing CDIs. Clostridium difficile can lead to various complications including septic shock and fulminant colitis that could prove to be lethal in these patients. Newer modalities of treatment have been developed including bezlotoxumab, a monoclonal antibody and fecal microbiota transplant. There have been studies showing asymptomatic carriers and drug resistance posing a major threat to the healthcare system. Newer treatment options are being studied to treat and prevent CDIs. This review will provide an insight into the current treatment modalities, prevention and newer modalities of treatment and challenges faced in the treatment of CDIs.
Collapse
Affiliation(s)
- Vignesh K Nagesh
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Hadrian Hoang-Vu Tran
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Daniel Elias
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Izage Kianifar Aguilar
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Tanni Sethi
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Aiswarya Menon
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Charlene Mansour
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Florchi Furman
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Kylie Tsotsos
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Talia Subar
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Auda Auda
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Aman Sidiqui
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Jevon Lamar
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Nikita Wadhwani
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Shraboni Dey
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Abraham Lo
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Adam Atoot
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Simcha Weissman
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Humberto Sifuentes
- Department of Gastroenterology, Augusta University, Augusta, GA 30912, United States
| | - Ayrton I Bangolo
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| |
Collapse
|
35
|
Herrera F, Torres D, Laborde A, Jordán R, Tula L, Mañez N, Pereyra ML, Suchowiercha N, Berruezo L, Gudiol C, Ibáñez MLG, Eusebio MJ, Lambert S, Barcán L, Rossi IR, Nicola F, Pennini M, Monge R, Blanco M, Visús M, Reynaldi M, Carbone R, Pasterán F, Corso A, Rapoport M, Carena AA. Seven-day antibiotic therapy for Enterobacterales bacteremia in high-risk neutropenic patients: toward a new paradigm. Eur J Clin Microbiol Infect Dis 2024; 43:1741-1751. [PMID: 38958809 DOI: 10.1007/s10096-024-04885-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE Data on short courses of antibiotic therapy for Enterobacterales bacteremia in high-risk neutropenic patients are limited. The aim of the study was to describe and compare the frequency of bacteremia relapse, 30-day overall and infection-related mortality, Clostridiodes difficile infection and length of hospital stay since bacteremia among those who received antibiotic therapy for 7 or 14 days. METHODS This is a multicenter, prospective, observational cohort study in adult high-risk neutropenic patients with hematologic malignancies or hematopoietic stem cell transplant and monomicrobial Enterobacterales bacteremia. They received appropriate empirical antibiotic therapy, had a clinical response within 7 days, and infection source control. Clinical, epidemiological and outcomes variables were compared based on 7 or 14 days of AT. RESULTS Two hundred patients were included (100, 7-day antibiotic therapy; 100, 14-day antibiotic therapy). Escherichia coli was the pathogen most frequently isolated (47.5%), followed by Klebsiella sp. (40.5%). Among those patients that received 7-day vs. 14-day antibiotic course, a clinical source of bacteremia was found in 54% vs. 57% (p = 0.66), multidrug-resistant Enterobacterales isolates in 28% vs. 30% (p = 0.75), and 40% vs. 47% (p = 0.31) received combined empirical antibiotic therapy. Overall mortality was 3% vs. 1% (p = 0.62), in no case related to infection; bacteremia relapse was 7% vs. 2% (p = 0.17), and length of hospital stay since bacteremia had a median of 9 days (IQR: 7-15) vs. 14 days (IQR: 13-22) (p = < 0.001). CONCLUSIONS These data suggest that seven-day antibiotic therapy might be adequate for patients with high-risk neutropenia and Enterobacterales bacteremia, who receive appropriate empirical therapy, with clinical response and infection source control.
Collapse
Affiliation(s)
- Fabián Herrera
- Infectious Diseases Section, Internal Medicine Department, Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Av. Galván 4102 (C1431), Buenos Aires, Argentina.
| | - Diego Torres
- Infectious Diseases Section, Internal Medicine Department, Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Av. Galván 4102 (C1431), Buenos Aires, Argentina
| | - Ana Laborde
- Infectious Diseases Service, Fundación Para Combatir La Leucemia (FUNDALEU), Buenos Aires, Argentina
| | - Rosana Jordán
- Infectious Diseases Service, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Lucas Tula
- Infectious Diseases Service, Hospital de Alta Complejidad El Cruce, Buenos Aires, Argentina
| | - Noelia Mañez
- Infectious Diseases Section, Internal Medicine Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - María Laura Pereyra
- Infectious Diseases Service, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Nadia Suchowiercha
- Infectious Diseases Service, Hospital Interzonal General de Agudos (HIGA) Gral. San Martín de La Plata, Buenos Aires, Argentina
| | - Lorena Berruezo
- Infectious Diseases Service, Hospital Interzonal General de Agudos (HIGA), Prof. Dr. Rodolfo Rossi de La Plata, Buenos Aires, Argentina
| | - Carlota Gudiol
- Infectious Diseases Department, Bellvitge University Hospital, IDIBELL, University of Barcelona, Barcelona, España
| | - María Luz González Ibáñez
- Infectious Diseases Service, Fundación Para Combatir La Leucemia (FUNDALEU), Buenos Aires, Argentina
| | - María José Eusebio
- Infectious Diseases Service, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Sandra Lambert
- Infectious Diseases Service, Hospital de Alta Complejidad El Cruce, Buenos Aires, Argentina
| | - Laura Barcán
- Infectious Diseases Section, Internal Medicine Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Inés Roccia Rossi
- Infectious Diseases Service, Hospital Interzonal General de Agudos (HIGA) Gral. San Martín de La Plata, Buenos Aires, Argentina
| | - Federico Nicola
- Microbiology Laboratory, Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Buenos Aires, Argentina
| | - Magdalena Pennini
- Microbiology Laboratory, Centro de Estudios Infectológicos (CEI) Dr. Stamboulian, Buenos Aires, Argentina
| | - Renata Monge
- Microbiology Service, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Miriam Blanco
- Microbiology Laboratory, Hospital de Alta Complejidad El Cruce, Buenos Aires, Argentina
| | - Mariángeles Visús
- Bacteriology Laboratory, Central Laboratory, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Reynaldi
- Microbiology Laboratory, Hospital Interzonal General de Agudos (HIGA), Gral. San Martín de La Plata, Buenos Aires, Argentina
| | - Ruth Carbone
- Bacteriology Laboratory, Hospital Interzonal General de Agudos (HIGA) Prof. Dr. Rodolfo Rossi de La Plata, Buenos Aires, Argentina
| | - Fernando Pasterán
- Antimicrobials Service, INEI-ANLIS Dr. Carlos Malbrán, Buenos Aires, Argentina
| | - Alejandra Corso
- Antimicrobials Service, INEI-ANLIS Dr. Carlos Malbrán, Buenos Aires, Argentina
| | - Melina Rapoport
- Antimicrobials Service, INEI-ANLIS Dr. Carlos Malbrán, Buenos Aires, Argentina
| | - Alberto Angel Carena
- Infectious Diseases Section, Internal Medicine Department, Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Av. Galván 4102 (C1431), Buenos Aires, Argentina
| |
Collapse
|
36
|
Jazmati N, Mischnik A, Kern WV, Behnke M, Chakraborty T, Dinkelacker A, Eisenbeis S, Falgenhauer J, Gastmeier P, Häcker G, Imirzalioglu C, Käding N, Kramme E, Peter S, Piepenbrock E, Rupp J, Schneider C, Schwab F, Seifert H, Tacconelli E, Trauth J, Biehl L, Walker SV, Rohde AM. Occurrence and trends of Clostridioides difficile infections in hospitalized patients: a prospective multi-centre cohort study in six German university hospitals, 2016-2020. J Hosp Infect 2024; 151:161-172. [PMID: 38969208 DOI: 10.1016/j.jhin.2024.06.007] [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: 04/16/2024] [Revised: 05/30/2024] [Accepted: 06/09/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND For Clostridioides difficile infections (CDIs) in Germany no longitudinal multi-centre studies with standardized protocols for diagnosing CDI are available. Recent evaluations of general surveillance databases in Germany indicate a downward trend in CDI rates. We aimed to describe the actual burden and trends of CDI in German university hospitals from 2016 to 2020. METHODS Our study was a prospective multi-centre study covering six German university hospitals. We report the data in total, stratified by year, by medical specialty as well as by CDI severity. Multi-variable regression analyses were performed to assess risk factors for severe CDI. RESULTS We registered 3780 CDI cases among 1,436,352 patients. The median length of stay (LOS) of CDI cases was 20 days (interquartile range 11-37) compared with a general LOS of 4.2 days. In-hospital all-cause mortality in CDI patients was 11.7% (N = 444/3780), while mortality attributed to CDI was 0.4% (N = 16/3761). CDI recurrence rate was comparatively low at 7.2%. The incidence density of severe healthcare-associated healthcare onset (HAHO)-CDI showed a significant decrease from 2.25/10,000 patient days (pd) in 2016 to 1.49/10,000 pd in 2020 (trend calculation P=0.032). CONCLUSIONS Compared with a European point-prevalence study in 2013/2014, where overall CDI incidence density was 11.2 cases/10,000 pd in Germany (EUCLID), we see in our study halved overall CDI rates of 5.6 cases/10,000 pd in 2020. Our study shows current data on the distribution of CDI cases in German university hospitals and thus provides international comparative data on the key indicators of CDI.
Collapse
Affiliation(s)
- N Jazmati
- German Centre for Infection Research (DZIF), Germany; Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Labor Dr. Wisplinghoff, Cologne, Germany.
| | - A Mischnik
- German Centre for Infection Research (DZIF), Germany; Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein/Campus Lübeck, Germany; Division of Infectious Diseases, Department of Medicine II, University Medical Centre and Faculty of Medicine, Albert-Ludwigs-University, Freiburg, Freiburg, Germany
| | - W V Kern
- Division of Infectious Diseases, Department of Medicine II, University Medical Centre and Faculty of Medicine, Albert-Ludwigs-University, Freiburg, Freiburg, Germany
| | - M Behnke
- German Centre for Infection Research (DZIF), Germany; Institute for Hygiene and Environmental Medicine, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - T Chakraborty
- German Centre for Infection Research (DZIF), Germany; Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - A Dinkelacker
- German Centre for Infection Research (DZIF), Germany; Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | - S Eisenbeis
- German Centre for Infection Research (DZIF), Germany; Division of Infectious Diseases, Department of Internal Medicine 1, University Hospital Tübingen, Tübingen, Germany
| | - J Falgenhauer
- German Centre for Infection Research (DZIF), Germany; Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - P Gastmeier
- German Centre for Infection Research (DZIF), Germany; Institute for Hygiene and Environmental Medicine, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - G Häcker
- German Centre for Infection Research (DZIF), Germany; Institute for Medical Microbiology and Hygiene, University Medical Centre Freiburg, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - C Imirzalioglu
- German Centre for Infection Research (DZIF), Germany; Institute of Medical Microbiology, Justus Liebig University Giessen, Germany
| | - N Käding
- German Centre for Infection Research (DZIF), Germany; Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein/Campus Lübeck, Germany
| | - E Kramme
- German Centre for Infection Research (DZIF), Germany; Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein/Campus Lübeck, Germany
| | - S Peter
- German Centre for Infection Research (DZIF), Germany; Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | - E Piepenbrock
- German Centre for Infection Research (DZIF), Germany; Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - J Rupp
- German Centre for Infection Research (DZIF), Germany; Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein/Campus Lübeck, Germany
| | - C Schneider
- German Centre for Infection Research (DZIF), Germany; Institute for Medical Microbiology and Hygiene, University Medical Centre Freiburg, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - F Schwab
- German Centre for Infection Research (DZIF), Germany; Institute for Hygiene and Environmental Medicine, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - H Seifert
- German Centre for Infection Research (DZIF), Germany; Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - E Tacconelli
- German Centre for Infection Research (DZIF), Germany; Division of Infectious Diseases, Department of Internal Medicine 1, University Hospital Tübingen, Tübingen, Germany
| | - J Trauth
- German Centre for Infection Research (DZIF), Germany; Department of Internal Medicine (Infectiology), Uniklinikum Giessen, Germany
| | - L Biehl
- German Centre for Infection Research (DZIF), Germany; Department I of Internal Medicine, University Hospital of Cologne, Germany
| | - S V Walker
- German Centre for Infection Research (DZIF), Germany; Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; RKH Regionale Kliniken Holding und Services GmbH, Institute for Clinical Microbiology and Hospital Hygiene, Hospital Ludwigsburg, Germany
| | - A M Rohde
- German Centre for Infection Research (DZIF), Germany; Institute for Hygiene and Environmental Medicine, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| |
Collapse
|
37
|
Nabukhotna K, Kordus SL, Shupe JA, Cano Rodríguez R, Smith A, Bohannon JK, Washington MK, Lacy DB. Purified CDT toxins and a clean deletion within the CDT locus provide novel insights into the contribution of binary toxin in cellular inflammation and Clostridioides difficile infection. PLoS Pathog 2024; 20:e1012568. [PMID: 39298531 PMCID: PMC11444381 DOI: 10.1371/journal.ppat.1012568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/01/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
Clostridioides difficile is a spore-forming pathogen and the most common cause of healthcare-associated diarrhea and colitis in the United States. Besides producing the main virulence factors, toxin A (TcdA) and toxin B (TcdB), many of the common clinical strains encode the C. difficile transferase (CDT) binary toxin. The role of CDT in the context of C. difficile infection (CDI) is poorly understood. Inflammation is a hallmark of CDI and multiple mechanisms of inflammasome activation have been reported for TcdA, TcdB, and the organism. Some studies have suggested that CDT contributes to this inflammation through a TLR2-dependent priming mechanism that leads to the suppression of protective eosinophils. Here, we show that CDT does not prime but instead activates the inflammasome in bone marrow-derived dendritic cells (BMDCs). In bone marrow-derived macrophages (BMDMs), the cell binding and pore-forming component of the toxin, CDTb, alone activates the inflammasome and is dependent on K+ efflux. The activation is not observed in the presence of CDTa and is not observed in BMDMs derived from Nlrp3-/- mice suggesting the involvement of the NLRP3 inflammasome. However, we did not observe evidence of CDT-dependent inflammasome priming or activation in vivo. Mice were infected with R20291 and an isogenic CRISPR/Cas9-generated R20291 ΔcdtB strain of C. difficile. While CDT contributes to increased weight loss and cecal edema at 2 days post infection, the relative levels of inflammasome-associated cytokines, IL-1β and IL-18, in the cecum and distal colon are unchanged. We also saw CDT-dependent weightloss in Nlrp3-/- mice, suggesting that the increased weightloss associated with the presence of CDT is not a result of NLRP3-dependent inflammasome activation. This study highlights the importance of studying gene deletions in the context of otherwise fully isogenic strains and the challenge of translating toxin-specific cellular responses into a physiological context, especially when multiple toxins are acting at the same time.
Collapse
Affiliation(s)
- Kateryna Nabukhotna
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Shannon L Kordus
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - John A Shupe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Rubén Cano Rodríguez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Anna Smith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Julia K Bohannon
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - D Borden Lacy
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
| |
Collapse
|
38
|
Kim J, Kim JS, Kim SH, Yoo S, Lee JK, Kim K. Deep learning-based prediction of Clostridioides difficile infection caused by antibiotics using longitudinal electronic health records. NPJ Digit Med 2024; 7:224. [PMID: 39181992 PMCID: PMC11344761 DOI: 10.1038/s41746-024-01215-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 08/02/2024] [Indexed: 08/27/2024] Open
Abstract
Clostridioides difficile infection (CDI) is a major cause of antibiotic-associated diarrhea and colitis. It is recognized as one of the most significant hospital-acquired infections. Although CDI can develop severe complications and spores of Clostridioides difficile can be transmitted by the fecal-oral route, CDI is occasionally overlooked in clinical settings. Thus, it is necessary to monitor high CDI risk groups, particularly those undergoing antibiotic treatment, to prevent complications and spread. We developed and validated a deep learning-based model to predict the occurrence of CDI within 28 days after starting antibiotic treatment using longitudinal electronic health records. For each patient, timelines of vital signs and laboratory tests with a 35-day monitoring period and a patient information vector consisting of age, sex, comorbidities, and medications were constructed. Our model achieved the prediction performance with an area under the receiver operating characteristic curve of 0.952 (95% CI: 0.932-0.973) in internal validation and 0.972 (95% CI: 0.968-0.975) in external validation. Platelet count and body temperature emerged as the most important features. The risk score, the output value of the model, exhibited a consistent increase in the CDI group, while the risk score in the non-CDI group either maintained its initial value or decreased. Using our CDI prediction model, high-risk patients requiring symptom monitoring can be identified. This could help reduce the underdiagnosis of CDI, thereby decreasing transmission and preventing complications.
Collapse
Affiliation(s)
- Junmo Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Joo Seong Kim
- Division of Gastroenterology, Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Republic of Korea
| | - Sae-Hoon Kim
- Division of Allergy & Clinical Immunology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sooyoung Yoo
- Office of eHealth Research and Businesses, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jun Kyu Lee
- Division of Gastroenterology, Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Republic of Korea.
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Yamada CH, Ortis GB, Buso GM, Martins TC, Zequinao T, Telles JP, Wollmann LC, Montenegro CDO, Dantas LR, Cruz JW, Tuon FF. Validation of Lyophilized Human Fecal Microbiota for the Treatment of Clostridioides difficile Infection: A Pilot Study with Pharmacoeconomic Analysis of a Middle-Income Country-Promicrobioma Project. Microorganisms 2024; 12:1741. [PMID: 39203583 PMCID: PMC11356882 DOI: 10.3390/microorganisms12081741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Clostridioides difficile infection (CDI) represents a prevalent and potentially severe health concern linked to the usage of broad-spectrum antibiotics. The aim of this study was to evaluate a new lyophilized product based on human fecal microbiota for transplant, including cost-benefit analysis in the treatment of recurrent or refractory CDI. METHODS The product for fecal microbiota transplant was obtained from two donors. Microbiological, viability, and genomic analysis were evaluated. After validation, a clinical pilot study including recurrent or refractory CDI with 24 patients was performed. Clinical response and 4-week recurrence were the outcome. Cost-benefit analysis compared the fecal microbiota transplant with conventional retreatment with vancomycin or metronidazole. RESULTS The microbiota for transplant presented significant bacterial viability, with and adequate balance of Firmicutes and Bacteroidetes. The clinical response with the microbiota transplant was 92%. In financial terms, estimated expenditure for CDI solely related to recurrence, based on stochastic modeling, totals USD 222.8 million per year in Brazil. CONCLUSIONS The lyophilized human fecal microbiota for transplant is safe and can be an important step for a new product with low cost, even with genomic sequencing. Fecal microbiota transplantation emerges as a more cost-effective alternative compared to antimicrobials in the retreatment of CDI.
Collapse
Affiliation(s)
- Carolina Hikari Yamada
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Gabriel Burato Ortis
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Gustavo Martini Buso
- School of Business, Pontifical Catholic University of Paraná, Rua Imaculada Conceição 1155, Curitiba 80215-901, PR, Brazil; (G.M.B.); (J.W.C.)
| | - Thalissa Colodiano Martins
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Tiago Zequinao
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Joao Paulo Telles
- Hospital Universitário Evangélico Mackenzie, Curitiba 80730-150, PR, Brazil; (J.P.T.); (L.C.W.)
| | | | - Carolina de Oliveira Montenegro
- School of Business, Pontifical Catholic University of Paraná, Rua Imaculada Conceição 1155, Curitiba 80215-901, PR, Brazil; (G.M.B.); (J.W.C.)
| | - Leticia Ramos Dantas
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - June Westarb Cruz
- School of Business, Pontifical Catholic University of Paraná, Rua Imaculada Conceição 1155, Curitiba 80215-901, PR, Brazil; (G.M.B.); (J.W.C.)
| | - Felipe Francisco Tuon
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| |
Collapse
|
40
|
Lee CC, Yan XZ, Wu HT, Ko WC, Tsai PJ, Hung YP. Potential effectiveness of parenteral nemonoxacin in the treatment of Clostridioides difficile infections: in vitro, ex vivo, and mouse studies. Front Microbiol 2024; 15:1418817. [PMID: 39228379 PMCID: PMC11368742 DOI: 10.3389/fmicb.2024.1418817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Introduction Antimicrobial therapy plays a crucial role in the management of CDI patients. However, the standard agent for treating CDIs is limited to oral fidaxomicin or vancomycin. For patients made nil by mouth, there is a clinically urgent and essential need to develop an intravenous antibiotic. Methods For C. difficile with the lowest MIC of nemonoxacin and vancomycin, the inhibitory effects were tested using the kinetic time-kill assay and ex vivo co-culture model. The effectiveness of nemonoxacin and vancomycin in inhibiting spore germination, the sporicidal activity, and the treatment of mice with CDIs were compared. Results For clinical isolates and laboratory strains, lower MICs of nemonoxacin against C. difficile than levofloxacin and ciprofloxacin were observed, even in those harboring point mutations in the quinolone-resistance determining region. Although nemonoxacin failed to suppress spore outgrowth and germination in C. difficile, it exhibited an effective inhibitory effect against C. difficile in the kinetic time-kill assay and the ex vivo co-culture model. Mice receiving intraperitoneal nemonoxacin had less weight loss, higher cecum weight, a longer colon length, and lower expression of the tcdB gene, compared with untreated mice. Notably, there were no significant differences observed in weight loss, cecum weight, colon length, or tcdB gene expression between mice treated with vancomycin and those treated with any dose of nemonoxacin. Similarly, no significant differences were found between mice receiving combination therapy of intraperitoneal nemonoxacin plus oral vancomycin and those treated with intraperitoneal nemonoxacin or oral vancomycin alone. Discussion The potential role of nemonoxacin, which can be administered parenterally, for treating CDIs was evidenced through the in vitro, ex vivo, and mouse models.
Collapse
Affiliation(s)
- Ching-Chi Lee
- Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Xiang-Zhe Yan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Tsung Wu
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Pin Hung
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| |
Collapse
|
41
|
Mitchell C, Keegan LT, Le TTT, Khader K, Beams A, Samore MH, Toth DJA. Importance of underlying mechanisms for interpreting relative risk of Clostridioides difficile infection among antibiotic-exposed patients in healthcare facilities. PLoS One 2024; 19:e0306622. [PMID: 39116083 PMCID: PMC11309424 DOI: 10.1371/journal.pone.0306622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/20/2024] [Indexed: 08/10/2024] Open
Abstract
Clostridioides difficile infection (CDI) is a significant public health threat, associated with antibiotic-induced disruption of the normally protective gastrointestinal microbiota. CDI is thought to occur in two stages: acquisition of asymptomatic colonization from ingesting C. difficile bacteria followed by progression to symptomatic CDI caused by toxins produced during C. difficile overgrowth. The degree to which disruptive antibiotic exposure increases susceptibility at each stage is uncertain, which might contribute to divergent published projections of the impact of hospital antibiotic stewardship interventions on CDI. Here, we model C. difficile transmission and CDI among hospital inpatients, including exposure to high-CDI-risk antibiotics and their effects on each stage of CDI epidemiology. We derive the mathematical relationship, using a deterministic model, between those parameters and observed equilibrium levels of colonization, CDI, and risk ratio of CDI among certain antibiotic-exposed patients relative to patients with no recent antibiotic exposure. We then quantify the sensitivity of projected antibiotic stewardship intervention impacts to alternate assumptions. We find that two key parameters, the antibiotic effects on susceptibility to colonization and to CDI progression, are not identifiable given the data frequently available. Furthermore, the effects of antibiotic stewardship interventions are sensitive to their assumed values. Thus, discrepancies between different projections of antibiotic stewardship interventions may be largely due to model assumptions. Data supporting improved quantification of mechanistic antibiotic effects on CDI epidemiology are needed to understand stewardship effects better.
Collapse
Affiliation(s)
- Christopher Mitchell
- Department of Mathematics, Tarleton State University, Stephenville, Texas, United States of America
| | - Lindsay T. Keegan
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States of America
- Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States of America
| | - Thuy T. T. Le
- Department of Health Management and Policy, School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Karim Khader
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States of America
- Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States of America
| | - Alexander Beams
- Department of Mathematics, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Matthew H. Samore
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States of America
- Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States of America
| | - Damon J. A. Toth
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States of America
- Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States of America
- Department of Mathematics, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
42
|
Angulo FJ, Furtado M, Gonzalez E, Zhang P, Kelly PH, Moïsi JC. Incidence of public health surveillance-reported Clostridioides difficile infections in thirteen countries worldwide: A narrative review. Anaerobe 2024; 88:102878. [PMID: 38909713 DOI: 10.1016/j.anaerobe.2024.102878] [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: 04/25/2024] [Revised: 06/06/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
Clostridioides difficile infection (CDI) is an important cause of morbidity and mortality worldwide. Data from public health surveillance systems are important for estimating country-level CDI burden. CDI surveillance can be population-based or hospital-based. Population-based surveillance results in overall estimates of CDI incidence (cases per 100,000 population-per-year), and hospital-based surveillance results in estimates of hospital-based CDI incidence (cases per 10,000 patient-days) or CDI admission rates (cases per 1,000 admissions). We sought to better understand temporal trends in CDI incidence reported in publicly available surveillance data worldwide and describe varying surveillance methods. We identified 13 countries in Europe, North America, and Oceania with publicly available population-based and/or hospital-based CDI surveillance data in online reports and/or dashboards. Additional countries in Europe, in particular, also conduct hospital-based CDI surveillance. Inconsistent CDI case definitions and surveillance approaches between countries limit the interpretability of multi-country comparisons. Nonetheless, publicly available CDI surveillance data enabled us to compare CDI incidence among countries with population-based and/or hospital-based surveillance systems and to describe trends in CDI incidence within countries over time. The highest CDI incidence is in the United States. While there have been recent declines in CDI incidence in all countries, the CDI burden remains high, and the need persists for CDI prevention strategies in communities and healthcare settings.
Collapse
Affiliation(s)
- Frederick J Angulo
- Vaccines & Antivirals, Chief Medical Affairs Office, Pfizer Biopharma, 500 Arcola Road, Collegeville, Pennsylvania 19426, USA.
| | - Melissa Furtado
- Medical Engagement and Impact, Chief Medical Affairs Office, Pfizer Biopharma, The Capital 1802/1901, G Block, Bandra-Kurla Complex, Mumbai 400051, Maharashtra, India
| | - Elisa Gonzalez
- Vaccines & Antivirals, Chief Medical Affairs Office, Pfizer Biopharma, 500 Arcola Road, Collegeville, Pennsylvania 19426, USA
| | - Pingping Zhang
- Medical Affairs Evidence Generation Statistics, Pfizer Research and Development, Collegeville, Pennsylvania, USA
| | - Patrick H Kelly
- Vaccines & Antivirals, Chief Medical Affairs Office, Pfizer Biopharma, 500 Arcola Road, Collegeville, Pennsylvania 19426, USA
| | - Jennifer C Moïsi
- Vaccines & Antivirals, Chief Medical Affairs Office, Pfizer Biopharma, 23-25 Avenue du Docteur Lannelongue, 75014 Paris, France
| |
Collapse
|
43
|
Ouyang M, Zou S, Cheng Q, Shi X, Zhao Y, Sun M. Vonoprazan-associated Clostridioides difficile infection: an analysis of the Japanese Adverse Drug Event Report and the FDA Adverse Event Reporting System. Ther Adv Drug Saf 2024; 15:20420986241260211. [PMID: 39091466 PMCID: PMC11292713 DOI: 10.1177/20420986241260211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/20/2024] [Indexed: 08/04/2024] Open
Abstract
Background Prolonged or excessive use of acid suppressants may increase the risk of Clostridioides difficile infection (CDI) by altering the intestinal microecosystem. Vonoprazan, a novel potassium-competitive acid blocker, exhibits a faster and more sustained acid-suppressive effect than proton pump inhibitors (PPIs). Therefore, vonoprazan may have a greater impact on the gut microbiota, potentially resulting in CDI. Objectives This study aimed to explore the potential relationship between acid suppressants and CDI by the Japan Adverse Drug Event Report (JADER) and the FDA Adverse Event Reporting System (FAERS) databases. Design A retrospective analysis of the JADER and FAERS databases was examined by disproportionality analysis. Methods We performed signal detection analyses of CDI induced by vonoprazan and PPIs using the JADER and FAERS databases. The association between acid suppressants and CDI was calculated using the reporting odds ratio (ROR) and corresponding 95% confidence interval (95% CI). When the lower limit of the 95% CI is exceeded by 1, the association is considered statistically significant. Results In the JADER database, the ROR (95% CI) for vonoprazan and PPIs based on suspect drug reports was 15.84 (12.23-20.50) and 2.51 (1.92-3.28), respectively. In the FAERS database, the ROR (95% CI) for vonoprazan and PPIs based on primary and secondary suspect drug reports was 11.50 (6.36-20.82) and 1.42 (1.34-1.51), respectively. Subgroup analysis showed that elderly patients aged 60 years and older were more strongly associated with CDI. The ROR (95% CI) for vonoprazan and PPIs in patients aged 60 years and older in the JADER database was 15.35 (11.59-20.33) and 1.65 (1.14-2.39), respectively. Similarly, the ROR (95% CI) for vonoprazan and PPIs in the FAERS database was 12.56 (6.26-25.20) and 1.43 (1.31-1.57), respectively. Excluding the effect of Helicobacter pylori (H. pylori) infection, the use of acid suppressants was still associated with CDI. Conclusion While signal detection analysis based on the JADER and FAERS databases could not establish causality, our study demonstrated that both vonoprazan and PPIs were significantly associated with CDI. Vonoprazan showed a stronger association with CDI in both databases.
Collapse
Affiliation(s)
- Mengling Ouyang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shupeng Zou
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qian Cheng
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xuan Shi
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yazheng Zhao
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Minghui Sun
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
44
|
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
|
45
|
Abouelkhair AA, Seleem MN. Exploring novel microbial metabolites and drugs for inhibiting Clostridioides difficile. mSphere 2024; 9:e0027324. [PMID: 38940508 PMCID: PMC11288027 DOI: 10.1128/msphere.00273-24] [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: 04/05/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024] Open
Abstract
Clostridioides difficile is an enteric pathogen that can cause a range of illnesses from mild diarrhea to pseudomembranous colitis and even death. This pathogen often takes advantage of microbial dysbiosis provoked by antibiotic use. With the increasing incidence and severity of infections, coupled with high recurrence rates, there is an urgent need to identify innovative therapies that can preserve the healthy state of the gut microbiota. In this study, we screened a microbial metabolite library against C. difficile. From a collection of 527 metabolites, we identified 18 compounds with no previously identified antimicrobial activity and metabolites that exhibited potent activity against C. difficile growth. Of these 18 hits, five drugs and three metabolites displayed the most potent anti-C. difficile activity and were subsequently assessed against 20 clinical isolates of C. difficile. These potent agents included ecteinascidin 770 (minimum inhibitory concentration against 50% of isolates [MIC50] ≤0.06 µg/mL); 8-hydroxyquinoline derivatives, such as broxyquinoline and choloroquinaldol (MIC50 = 0.125 µg/mL); ionomycin calcium salt, carbadox, and robenidine hydrochloride (MIC50 = 1 µg/mL); and dronedarone and milbemycin oxime (MIC50 = 4 µg/mL). Unlike vancomycin and fidaxomicin, which are the standard-of-care anti-C. difficile antibiotics, most of these metabolites showed robust bactericidal activity within 2-8 h with minimal impact on the growth of representative members of the normal gut microbiota. These results suggest that the drugs and microbial metabolite scaffolds may offer alternative avenues to address unmet needs in C. difficile disease prevention and treatment. IMPORTANCE The most frequent infection associated with hospital settings is Clostridioides difficile, which can cause fatal diarrhea and severe colitis, toxic megacolon, sepsis, and leaky gut. Those who have taken antibiotics for other illnesses that affect the gut's healthy microbiota are more susceptible to C. difficile infection (CDI). Recently, some reports showed higher recurrence rates and resistance to anti-C. difficile, which may compromise the efficacy of CDI treatment. Our study is significant because it is anticipated to discover novel microbial metabolites and drugs with microbial origins that are safe for the intestinal flora, effective against C. difficile, and reduce the risk of recurrence associated with CDI.
Collapse
Affiliation(s)
- Ahmed A. Abouelkhair
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Department of Bacteriology, Mycology, and Immunology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Mohamed N. Seleem
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|
46
|
Hunt A, Drwiega E, Wang Y, Danziger L. A review of fecal microbiota, live-jslm for the prevention of recurrent Clostridioides difficile infection. Am J Health Syst Pharm 2024; 81:e402-e411. [PMID: 38470061 DOI: 10.1093/ajhp/zxae066] [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: 03/09/2024] [Indexed: 03/13/2024] Open
Abstract
PURPOSE To review the composition, preparation, proposed mechanism of action, safety, efficacy, and current place in therapy of Rebyota (fecal microbiota, live-jslm). SUMMARY As the first agent in a new class of drugs, live biotherapeutic products (LBPs), fecal microbiota, live-jslm offers another therapeutic approach for the prevention of recurrent Clostridioides difficile infection (rCDI). LBPs are given following antibiotic therapy for C. difficile to reintroduce certain bacteria present in the normal microbiome, as a means to reconstitute the microbiome of infected individuals. This review provides a summary of phase 2 and 3 clinical trials, product information, discussion of data limitations, and recommendations for place in therapy. High efficacy rates compared to placebo with sustained response up to 24 months after administration have been reported. The majority of adverse events identified were mild to moderate without significant safety signals. CONCLUSION Fecal microbiota, live-jslm has consistently been shown in randomized trials to be safe and effective in reducing rCDI. Its approval marks the culmination of decades of work to identify, characterize, and refine the intestinal microbiome to create pharmaceutical products.
Collapse
Affiliation(s)
- Aaron Hunt
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| | - Emily Drwiega
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| | - Yifan Wang
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| | - Larry Danziger
- University of Illinois Chicago College of Pharmacy, Chicago, IL, USA
| |
Collapse
|
47
|
Long AE, Pitta D, Hennessy M, Indugu N, Vecchiarelli B, Luethy D, Aceto H, Hurcombe S. Assessment of fecal bacterial viability and diversity in fresh and frozen fecal microbiota transplant (FMT) product in horses. BMC Vet Res 2024; 20:306. [PMID: 38987780 PMCID: PMC11234551 DOI: 10.1186/s12917-024-04166-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Currently, lack of standardization for fecal microbiota transplantation (FMT) in equine practice has resulted in highly variable techniques, and there is no data on the bacterial metabolic activity or viability of the administered product. The objectives of this study were to compare the total and potentially metabolically active bacterial populations in equine FMT, and assess the effect of different frozen storage times, buffers, and temperatures on an equine FMT product. Fresh feces collected from three healthy adult horses was subjected to different storage methods. This included different preservation solutions (saline plus glycerol or saline only), temperature (-20 °C or -80 °C), and time (fresh, 30, 60, or 90 days). Samples underwent DNA extraction to assess total bacterial populations (both live and dead combined) and RNA extraction followed by reverse transcription to cDNA as a proxy to assess viable bacteria, then 16s rRNA gene amplicon sequencing using the V1-V2 region. RESULTS The largest difference in population indices and taxonomic composition at the genus level was seen when evaluating the results of DNA-based (total) and cDNA-based (potentially metabolically active) extraction method. At the community level, alpha diversity (observed species, Shannon diversity) was significantly decreased in frozen samples for DNA-based analysis (P < 0.05), with less difference seen for cDNA-based sequencing. Using DNA-based analysis, length of storage had a significant impact (P < 0.05) on the bacterial community profiles. For potentially metabolically active populations, storage overall had less of an effect on the bacterial community composition, with a significant effect of buffer (P < 0.05). Individual horse had the most significant effect within both DNA and cDNA bacterial communities. CONCLUSIONS Frozen storage of equine FMT material can preserve potentially metabolically active bacteria of the equine fecal microbiome, with saline plus glycerol preservation more effective than saline alone. Larger studies are needed to determine if these findings apply to other individual horses. The ability to freeze FMT material for use in equine patients could allow for easier clinical use of fecal transplant in horses with disturbances in their intestinal microbiome.
Collapse
Affiliation(s)
- Alicia E Long
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA.
| | - Dipti Pitta
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Meagan Hennessy
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Nagaraju Indugu
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Bonnie Vecchiarelli
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Daniela Luethy
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Helen Aceto
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Samuel Hurcombe
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
- Veterinary Innovative Partners, New York, NY, USA
| |
Collapse
|
48
|
Kim HH, Park SS, Kim BC, Han KS, Kim B, Hong CW, Sohn DK, You K, Lee DW, Park SC. Treatment for appendicitis in cancer patients on chemotherapy: a retrospective cohort study. Ann Surg Treat Res 2024; 107:1-7. [PMID: 38978688 PMCID: PMC11227919 DOI: 10.4174/astr.2024.107.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 07/10/2024] Open
Abstract
Purpose Whether to perform surgery or conservatively manage appendicitis in immunosuppressed patients is a concern for clinicians. This study aimed to compare the outcomes of these 2 treatment options for appendicitis in patients with cancer undergoing chemotherapy. Methods This retrospective study included 206 patients with cancer who were diagnosed with acute appendicitis between August 2001 and December 2021. Among them, patients who received chemotherapy within 1 month were divided into surgical and conservative groups. We evaluated the outcomes, including treatment success within 1 year, 1-year recurrence, and the number of days from the diagnosis of appendicitis to chemotherapy restart, between the 2 groups. Results Among the 206 patients with cancer who were diagnosed with acute appendicitis, 78 received chemotherapy within 1 month. The patients were divided into surgery (n = 63) and conservative (n = 15) groups. In the surgery group, the duration of antibiotic therapy (7.0 days vs. 16.0 days, P < 0.001) and length of hospital stay (8.0 days vs. 27.5 days, P = 0.002) were significantly shorter than conservative groups. The duration from the diagnosis of appendicitis to the restart of chemotherapy was shorter in the surgery group (20.8 ± 15.1 days vs. 35.2 ± 28.2 days, P = 0.028). The treatment success rate within 1 year was higher in the surgery group (100% vs. 33.3%, P < 0.001). Conclusion Surgical treatment showed a significantly higher success rate than conservative treatment for appendicitis in patients less than 1 month after chemotherapy. Further prospective studies will be needed to clinically determine treatment options.
Collapse
Affiliation(s)
- Hyung Hwan Kim
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Sung Sil Park
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Byung Chang Kim
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Kyung Su Han
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Bun Kim
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Chang Won Hong
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Dae Kyung Sohn
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Kiho You
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Dong Woon Lee
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | - Sung Chan Park
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| |
Collapse
|
49
|
Yadegar A, Bar-Yoseph H, Monaghan TM, Pakpour S, Severino A, Kuijper EJ, Smits WK, Terveer EM, Neupane S, Nabavi-Rad A, Sadeghi J, Cammarota G, Ianiro G, Nap-Hill E, Leung D, Wong K, Kao D. Fecal microbiota transplantation: current challenges and future landscapes. Clin Microbiol Rev 2024; 37:e0006022. [PMID: 38717124 PMCID: PMC11325845 DOI: 10.1128/cmr.00060-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
SUMMARYGiven the importance of gut microbial homeostasis in maintaining health, there has been considerable interest in developing innovative therapeutic strategies for restoring gut microbiota. One such approach, fecal microbiota transplantation (FMT), is the main "whole gut microbiome replacement" strategy and has been integrated into clinical practice guidelines for treating recurrent Clostridioides difficile infection (rCDI). Furthermore, the potential application of FMT in other indications such as inflammatory bowel disease (IBD), metabolic syndrome, and solid tumor malignancies is an area of intense interest and active research. However, the complex and variable nature of FMT makes it challenging to address its precise functionality and to assess clinical efficacy and safety in different disease contexts. In this review, we outline clinical applications, efficacy, durability, and safety of FMT and provide a comprehensive assessment of its procedural and administration aspects. The clinical applications of FMT in children and cancer immunotherapy are also described. We focus on data from human studies in IBD in contrast with rCDI to delineate the putative mechanisms of this treatment in IBD as a model, including colonization resistance and functional restoration through bacterial engraftment, modulating effects of virome/phageome, gut metabolome and host interactions, and immunoregulatory actions of FMT. Furthermore, we comprehensively review omics technologies, metagenomic approaches, and bioinformatics pipelines to characterize complex microbial communities and discuss their limitations. FMT regulatory challenges, ethical considerations, and pharmacomicrobiomics are also highlighted to shed light on future development of tailored microbiome-based therapeutics.
Collapse
Affiliation(s)
- Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Haggai Bar-Yoseph
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tanya Marie Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Sepideh Pakpour
- School of Engineering, Faculty of Applied Sciences, UBC, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Andrea Severino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ed J Kuijper
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Wiep Klaas Smits
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Sukanya Neupane
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Sadeghi
- School of Engineering, Faculty of Applied Sciences, UBC, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Estello Nap-Hill
- Department of Medicine, Division of Gastroenterology, St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dickson Leung
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Wong
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
50
|
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
|