1
|
Xaplanteri P, Oikonomopoulou C, Xini C, Potsios C. Community-Acquired Clostridioides difficile Infection: The Fox Among the Chickens. Int J Mol Sci 2025; 26:4716. [PMID: 40429858 PMCID: PMC12112421 DOI: 10.3390/ijms26104716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/03/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Clostridioides difficile infection (CDI) appears mainly as nosocomial antibiotic-associated diarrhea, and community-acquired infection is increasingly being recognized. The threshold of asymptomatic colonization and the clinical manifestation of CDI need further elucidation. Community-acquired CDI (CA-CDI) should be considered when the disease commences within 48 h of admission to hospital or more than 12 weeks after discharge. Although CDI is not established as a food-borne or zoonotic disease, some data support that direction. The spores' ability to survive standard cooking procedures and on abiotic surfaces, the formation of biofilms, and their survival within biofilms of other bacteria render even a low number of spores capable of food contamination and spread. Adequate enumeration methods for detecting a low number of spores in food have not been developed. Primary care physicians should take CA-CDI into consideration in the differential diagnosis of diarrhea, as there is a thin line between colonization and infection. In patients diagnosed with inflammatory bowel disease and other comorbidities, C. difficile can be the cause of recurrent disease and should be included in the estimation of diarrhea and worsening colitis symptoms. In the community setting, it is difficult to distinguish asymptomatic carriage from true infection. For asymptomatic carriage, antibiotic therapy is not suggested but contact isolation and hand-washing practices are required. Primary healthcare providers should be vigilant and implement infection control policies for the prevention of C. difficile spread.
Collapse
Affiliation(s)
- Panagiota Xaplanteri
- Department of Microbiology, General Hospital of Eastern Achaia, 25100 Aigio, Greece
| | | | - Chrysanthi Xini
- Department of Microbiology, Attikon University General Hospital, 12462 Athens, Greece;
| | - Charalampos Potsios
- Department of Internal Medicine, University General Hospital of Patras, 26504 Patras, Greece;
| |
Collapse
|
2
|
Litvinov E, Litvinov A. Impact of Clindamycin on the Oral-Gut Axis: Gastrointestinal Side Effects and Clostridium difficile Infection in 45 Patients. Cureus 2024; 16:e75381. [PMID: 39781176 PMCID: PMC11710861 DOI: 10.7759/cureus.75381] [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] [Accepted: 12/09/2024] [Indexed: 01/12/2025] Open
Abstract
Introduction The use of antibiotics such as oral clindamycin has been effective in treating bacterial infections. However, this medication often comes with significant side effects, particularly those affecting the gastrointestinal (GI) system. This study aims to evaluate the impact of different doses of clindamycin on GI health, specifically examining side effects like stomach upset, diarrhea duration, stomach pain, and recovery time. Given that clindamycin is frequently prescribed, understanding its impact on the oral-gut axis is critical to optimizing antibiotic therapy and reducing adverse events. Background Clindamycin, a lincosamide antibiotic, is widely used to treat a variety of bacterial infections. It acts by inhibiting bacterial protein synthesis but, like many antibiotics, also has unintended consequences for human gut health. The oral-gut axis represents a complex connection where antibiotics, such as clindamycin, can significantly alter the microbiota, leading to imbalances that manifest as diarrhea, abdominal pain, and other digestive issues. This study aims to explore these effects in depth by comparing two common doses of clindamycin, 300 mg versus 600 mg, and the impact of each dose on the severity and duration of GI side effects. Materials and methods This study involves 45 patients prescribed clindamycin for various bacterial infections. The patients were evaluated in two groups: 22 patients who received 300 mg and 23 patients who received 600 mg. Treatment duration ranged from seven to 10 days. Data collection focused on patient-reported symptoms, including the presence and duration of stomach upset, the length of diarrhea episodes, the persistence of stomach pain, and the overall recovery time. Statistical analysis included independent t-tests to compare symptom severity between the groups and chi-squared tests to assess differences in the incidence of side effects, while regression analysis was used to examine predictors of prolonged GI symptoms. Results The results of the study showed that 98% of patients experienced some side effects from oral clindamycin. Among those receiving the 600 mg dose, the frequency and severity of side effects were significantly higher compared to the 300 mg group. Specifically, the average duration of diarrhea in the 600 mg group was five days, compared to three days in the 300 mg group. Similarly, the average length of stomach pain in the higher dose group was seven days, compared to four days for those taking the lower dose. Chi-squared analysis indicated a significant association between the higher dose and increased incidence of GI symptoms. Regression analysis further showed that the 600 mg dose was a significant predictor of prolonged GI disturbances, underscoring a dose-dependent relationship. Conclusion The findings of this case study highlight that oral clindamycin, particularly at higher doses, is associated with increased GI side effects, including prolonged diarrhea and stomach pain. Almost all patients experienced side effects, with those on the 600 mg dose suffering more severe and prolonged symptoms compared to those on the 300 mg dose. The results suggest avoiding the prescription of oral clindamycin unless absolutely necessary, to reduce adverse outcomes and improve compliance. It is recommended to prioritize first-line antibiotics and reserve oral clindamycin as a secondary option. Further research is needed to investigate strategies for prescribing.
Collapse
Affiliation(s)
| | - Alan Litvinov
- Private Practice and Research, American Dental Association, Penfield, USA
| |
Collapse
|
3
|
Duo H, Yang Y, Zhang G, Chen Y, Cao Y, Luo L, Pan H, Ye Q. Comparative effectiveness of treatments for recurrent Clostridioides difficile infection: a network meta-analysis of randomized controlled trials. Front Pharmacol 2024; 15:1430724. [PMID: 39484168 PMCID: PMC11525118 DOI: 10.3389/fphar.2024.1430724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Background Clostridioides difficile infection (CDI) is the most common cause of healthcare-associated infectious diarrhea. A major clinical challenge is recurrent CDI (rCDI) without effective standard drug-based therapy. Additionally, a comprehensive comparison of various therapy effectiveness in rCDI patients is still under investigation. Methods A Bayesian network meta-analysis (NMA) of randomized control trials up to March 2024 was performed to investigate the efficacy of rCDI interventions. Results Seventeen trials were included, comprising 4,148 CDI patients with ten interventions, including fecal microbiota transplantation (FMT) by lower gastrointestinal (LGI), FMT by upper gastrointestinal (UGI), Autologous FMT (AFMT), vancomycin + FMT, vancomycin, placebo, fidaxomicin, Vowst (SER109), Rebyota (RBX2660), and monoclonal antibody. NMA showed that FMT by LGI had the highest efficacy in treating rCDIs with an odds ratio (95% confidence interval) of 32.33 (4.03, 248.69) compared with placebo. FMT by UGI also showed high efficacy, whereas the efficacy comparison between FMT by LGI and UGI was not statistically significant (ORs) (95% CI), 1.72 (0.65, 5.21). The rankogram and surface under the cumulative ranking curve (SUCRA) also showed FMT by LGI ranked at the top and FMT by UGI ranked second in the curative effect. Conclusion NMA demonstrates FMT's significant efficacy in rCDI management, regardless of administration route (lower or upper gastrointestinal). Despite its significant benefits, FMT's safety is a concern due to the lack of standardized FDAcompliant manufacturing and oversight. Microbiota-based therapies also exhibit potential. However, limited research mandates further clinical exploration. Antibiotics, in contrast, display comparatively reduced efficacy in rCDI, potentially linked to disruptions in native gut microflora balance. Systematic Review https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=368435, Identifier CRD42022368435.
Collapse
Affiliation(s)
- Hong Duo
- Hubei Key Laboratory of Medical Technology on Transplantation, National Quality Control Center for Donated Organ Procurement, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| | - Yanwei Yang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Guqing Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingxin Chen
- Global Health Institute, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yumeng Cao
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linjie Luo
- Department of Experimental Radiation Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Huaqin Pan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplantation Intensive Care Unit, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Qifa Ye
- Hubei Key Laboratory of Medical Technology on Transplantation, National Quality Control Center for Donated Organ Procurement, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
4
|
Lou K, Luo H, Jiang X, Feng S. Applications of emerging extracellular vesicles technologies in the treatment of inflammatory diseases. Front Immunol 2024; 15:1364401. [PMID: 38545101 PMCID: PMC10965547 DOI: 10.3389/fimmu.2024.1364401] [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: 01/02/2024] [Accepted: 03/04/2024] [Indexed: 04/17/2024] Open
Abstract
The emerging extracellular vesicles technologies is an advanced therapeutic approach showing promising potential for addressing inflammatory diseases. These techniques have been proven to have positive effects on immune modulation and anti-inflammatory responses. With these advancements, a comprehensive review and update on the role of extracellular vesicles in inflammatory diseases have become timely. This review aims to summarize the research progress of extracellular vesicle technologies such as plant-derived extracellular vesicles, milk-derived extracellular vesicles, mesenchymal stem cell-derived extracellular vesicles, macrophage-derived extracellular vesicles, etc., in the treatment of inflammatory diseases. It elucidates their potential significance in regulating inflammation, promoting tissue repair, and treating diseases. The goal is to provide insights for future research in this field, fostering the application and development of extracellular vesicle technology in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Kecheng Lou
- Department of Urology, Lanxi People’s Hospital, Jinhua, Zhejiang, China
| | - Hui Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinghua Jiang
- Department of Urology, Jingdezhen Second People’s Hospital, Jingdezhen, Jiangxi, China
| | - Shangzhi Feng
- Department of Urology, Jiujiang University Clinic College/Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
5
|
Chang KC, Nagarajan N, Gan YH. Short-chain fatty acids of various lengths differentially inhibit Klebsiella pneumoniae and Enterobacteriaceae species. mSphere 2024; 9:e0078123. [PMID: 38305176 PMCID: PMC10900885 DOI: 10.1128/msphere.00781-23] [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: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
The gut microbiota is inextricably linked to human health and disease. It can confer colonization resistance against invading pathogens either through niche occupation and nutrient competition or via its secreted metabolites. Short-chain fatty acids (SCFA) are the primary metabolites in the gut as a result of dietary fiber fermentation by the gut microbiota. In this work, we demonstrate that the interaction of single-species gut commensals on solid media is insufficient for pathogen inhibition, but supernatants from monocultures of these commensal bacteria enriched in acetate confer inhibition against anaerobic growth of the enteric pathogen Klebsiella pneumoniae. The three primary SCFAs (acetate, propionate, and butyrate) strongly inhibit the intestinal commensal Escherichia coli Nissle as well as a panel of enteric pathogens besides K. pneumoniae at physiological pH of the cecum and ascending colon. This inhibition was significantly milder on anaerobic gut commensals Bacteroides thetaiotaomicron and Bifidobacterium adolescentis previously demonstrated to be associated with microbiota recovery after antibiotic-induced dysbiosis. We describe a general suppression of bacterial membrane potential by these SCFAs at physiological cecum and ascending colonic pH. Furthermore, the strength of bacterial inhibition increases with increasing alkyl chain length. Overall, the insights gained in this study shed light on the potential therapeutic use of SCFAs for conferring colonization resistance against invading pathogens in a dysbiotic gut.IMPORTANCERising antimicrobial resistance has made treatment of bacterial infections increasingly difficult. According to the World Health Organization, it has become a burgeoning threat to hospital and public health systems worldwide. This threat is largely attributed to the global rise of carbapenem-resistant Enterobacteriaceae in recent years, with common hospital-acquired pathogens growing increasingly resistant to last-line antibiotics. Antibiotics disrupt the homeostatic balance of the gut microbiota, resulting in the loss of colonization resistance against enteric pathogens. This work describes the ability of short-chain fatty acids (SCFAs) produced by gut microbiota to be effective against a wide panel of enteric pathogens without major impact on common gut commensal species. We also demonstrate a previously undescribed link between alkyl chain length and antibacterial effects of SCFAs. SCFAs, thus, hold promise as an alternative therapeutic option leveraging on the antimicrobial activity of these endogenously produced gut metabolites without disrupting gut microbiota homeostasis.
Collapse
Affiliation(s)
- Kai Chirng Chang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Niranjan Nagarajan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yunn-Hwen Gan
- Department of Biochemistry, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Wu Z, Xu Q, Gu S, Chen Y, Lv L, Zheng B, Wang Q, Wang K, Wang S, Xia J, Yang L, Bian X, Jiang X, Zheng L, Li L. Akkermansia muciniphila Ameliorates Clostridioides difficile Infection in Mice by Modulating the Intestinal Microbiome and Metabolites. Front Microbiol 2022; 13:841920. [PMID: 35663882 PMCID: PMC9159907 DOI: 10.3389/fmicb.2022.841920] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Clostridioides difficile is a common cause of nosocomial infection. Antibiotic-induced dysbiosis in the intestinal microbiota is a core cause of C. difficile infection (CDI). Akkermansia muciniphila plays an active role in maintaining gastrointestinal balance and might offer the protective effects on CDI as probiotics. Here, we investigated the effects and mechanisms of A. muciniphila on CDI. C57BL/6 mice (n = 29) were administered A. muciniphila Muc T (3 × 109 CFUs, 0.2 mL) or phosphate-buffered saline (PBS) by oral gavage for 2 weeks. Mice were pretreated with an antibiotic cocktail and subsequently challenged with the C. difficile strain VPI 10463. A. muciniphila treatment prevented weight loss in mice and reduced the histological injury of the colon. And it also alleviated inflammation and improved the barrier function of the intestine. The administration effects of A. muciniphila may be associated with an increase in short-chain fatty acid production and the maintenance of bile acids' steady-state. Our results provide evidence that administration of A. muciniphila to CDI mice, with an imbalance in the microbial community structure, lead to a decrease in abundance of members of the Enterobacteriaceae and Enterococcaceae. In short, A. muciniphila shows a potential anti-CDI role by modulating gut microbiota and the metabolome.
Collapse
Affiliation(s)
- Zhengjie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaomai Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Silan Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaicen Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuting Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyuan Bian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianwan Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lisi Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Bacterial Research Platform, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| |
Collapse
|
7
|
Gupta A, Savanti F, Singh B, Sachdev P, Raj D, Garg I, Aruwani SK, Shaukat F. Risk Factors Associated With Clostridium difficile-Associated Diarrhea. Cureus 2021; 13:e18115. [PMID: 34692326 PMCID: PMC8527547 DOI: 10.7759/cureus.18115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2021] [Indexed: 11/27/2022] Open
Abstract
Introduction: Recent years have been alarming due to the sudden, dramatic rise in the incidence of Clostridium difficile infection (CDI). Identifying and addressing the risk factors associated with CDI will help in reducing the incidence of infection and associated complications. Methods: This case-control study was conducted in a tertiary care hospital in Pakistan from June 2020 to March 2021, in which 200 patients diagnosed with Clostridium difficile-associated diarrhea (CDAD) were enrolled in the study. CDAD was diagnosed based on clinical symptoms and stool enzyme immunoassay. Another 200 participants without a diagnosis of CDAD were enrolled from the outpatient department as a control group. Participants were enrolled after seeking informed consent. Results: In patients older than 65, risk of CDI was higher compared to participants lower than 65 years old (15.5% vs. 8.0%; p value: 0.02). Hospitalization (25.5% vs. 6.0%; p value < 0.0001), the use of proton pump inhibitors in last 30 days (23.0% vs. 10.5%; p value: 0.001) , and use of antibiotics in the last 30 days (36.0% vs. 10.5%; p value < 0.0001) were significantly higher in participants with CDI. Conclusion: Hospitalization, the usage of proton pump inhibitors, and antibiotics in the last 30 days were significantly associated with CDI. A higher incidence of CDI was associated with risk factors like increased body mass index, diabetes, chronic kidney disease, and malignancy.
Collapse
Affiliation(s)
- Aarzoo Gupta
- Internal Medicine, Safdarjung Hospital, Faridabad, IND
| | - Fnu Savanti
- Internal Medicine, Liaquat National Hospital and Medical College, Karachi, PAK
| | - Balvender Singh
- Internal Medicine, Ghulam Mohammad Mahar Medical College, Sukkur, PAK
| | - Priyanka Sachdev
- Internal Medicine, Liaquat University of Medical and Health Sciences, Jamshoro, PAK
| | - Deepak Raj
- Internal Medicine, Liaquat University of Medical and Health Sciences, Jamshoro, PAK
| | - Ishan Garg
- Internal Medicine, Ross University School of Medicine, Miami, USA
| | - Suraj K Aruwani
- Internal Medicine, Jinnah Sindh Medical University, Karachi, PAK
| | - Faizan Shaukat
- Internal Medicine, Dow University of Health Sciences, Karachi, PAK
| |
Collapse
|
8
|
Yun JH, Park GE, Ki HK. Correlation between antibiotic consumption and the incidence of healthcare facility-onset Clostridioides difficile infection: a retrospective chart review and analysis. Antimicrob Resist Infect Control 2021; 10:117. [PMID: 34362442 PMCID: PMC8348999 DOI: 10.1186/s13756-021-00986-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 07/14/2021] [Indexed: 01/05/2023] Open
Abstract
Background Healthcare facility-onset Clostridioides difficile infection is the leading cause of antibiotic-associated diarrhea, and is associated with morbidity and mortality. The use of antibiotics is an important risk factor for healthcare facility-onset C. difficile infection. We evaluated the correlation between the incidence of healthcare facility-onset C. difficile infection and antibiotic consumption, according to antibiotic class. Methods Patients with healthcare facility-onset C. difficile infection from January 2017 to December 2018 at Konkuk University Medical Center (a tertiary medical center) were included. We evaluated changes in the incidence of healthcare facility-onset C. difficile infection and antibiotic consumption. The correlation between the incidence of healthcare facility-onset C. difficile infection and antibiotic consumption was evaluated two ways: without a time interval and with 1-month interval matching. Results A total of 446 episodes of healthcare facility-onset C. difficile infection occurred during the study period. The incidence of healthcare facility-onset C. difficile infection was 9.3 episodes per 10,000 patient-days, and increased significantly. We observed an increase in the consumption of β-lactam/β-lactamase inhibitors, and a decrease in the consumption of other classes of antibiotics, with a significant decrease in the consumption of fluoroquinolones, glycopeptides, and clindamycin (P = 0.01, P < 0.001, and P = 0.001, respectively). The consumption of β-lactam/β-lactamase inhibitors was independently correlated with the incidence of healthcare facility-onset C. difficile infection in the analysis without a time interval. When the analysis was conducted with 1-month interval matching, glycopeptide consumption was independently associated with the incidence of healthcare facility-onset C. difficile infection. Conclusions Despite the reduction in fluoroquinolone and clindamycin consumption, the incidence of healthcare facility-onset C. difficile infection increased during the study period, and was correlated with increased consumption of β-lactam/β-lactamase inhibitors. Reduced consumption of specific antibiotics may be insufficient to reduce the incidence of healthcare facility-onset C. difficile infection. Supplementary Information The online version contains supplementary material available at 10.1186/s13756-021-00986-9.
Collapse
Affiliation(s)
- Ji Hyun Yun
- Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, Konkuk University, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ga Eun Park
- Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, Konkuk University, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Hyun Kyun Ki
- Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, Konkuk University, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
9
|
Chaar A, Feuerstadt P. Evolution of clinical guidelines for antimicrobial management of Clostridioides difficile infection. Therap Adv Gastroenterol 2021; 14:17562848211011953. [PMID: 33995583 PMCID: PMC8111514 DOI: 10.1177/17562848211011953] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/31/2021] [Indexed: 02/04/2023] Open
Abstract
Clostridioides difficile infection (CDI) has been an epidemic for many years. Our biggest challenge in treating CDI is preventing recurrence, which is seen in approximately 25% of patients with initial infection and in 40-60% of those with subsequent episodes. Given the major disease burden of this infection, appropriate data-driven treatment remains essential. Clinical treatment guidelines provide an unbiased critical analysis of the literature, integrating the quality of the available data to make recommendations. As CDI has been evolving and more research has become available, the frequency of guideline issue from various global societies has increased, as has the detail of the recommendations to fit more relevant clinical scenarios. In this review, we will discuss clinical guideline recommendations over three time periods: The Initial Guidelines 1995-1997, The Second Wave 2009-2013, and The Modern Era 2014-present. We see the changing recommendations from metronidazole or vancomycin for initial infection during earlier times to preferential treatment with fidaxomicin within the Infectious Diseases Society of America (IDSA) and Society of Healthcare Epidemiology of America (SHEA) joint guidelines provisional update in late 2020. The recommended treatments for first recurrence were initially with the same antimicrobial as the first episode but have since changed to having multiple options for one or more recurrences. We have also seen the addition of immune boosting treatments, including fecal microbiota transplantation (FMT)/microbiota restoration therapy (MRT) and bezlotoxumab in the more modern recommendations. As the guidelines are evolving with the times, it remains important to understand the differences among them so we can apply this information clinically and optimize patient outcomes.
Collapse
Affiliation(s)
- Abdelkader Chaar
- Section of Internal Medicine, Yale-New Haven Hospital, New Haven, CT, USA
| | | |
Collapse
|
10
|
Johnson SH, Waisbren SJ. Physician Responsiveness to Positive Blood Culture Results at the Minneapolis Veterans Affairs Hospital-Is Anyone Paying Attention? Fed Pract 2021; 38:128-135. [PMID: 33859464 DOI: 10.12788/fp.0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Patients presenting with suspected infections are typically placed on empiric broad-spectrum antibiotics. With mounting evidence supporting the efficacy of using the narrowest spectrum of antimicrobial therapy to cover the suspected pathogen, current guidelines recommend decreasing the breadth of coverage in response to culture results both in relation to microbe identification and antibiotic sensitivity. Methods A retrospective chart review of electronic health records at the Minneapolis Veterans Affairs Medical Center (VAMC) in Minnesota was performed for 208 positive blood cultures with antibiotic spectrum analysis from July 1, 2015 to June 30, 2016. The time of reporting for pathogen identification and subsequent pathogen susceptibilities were compared to the time at which any alterations to antibiotic coverage were made. The breadth of antibiotic coverage was recorded using a nonlinear spectrum score. The use of this score allowed for the reliable classification of antibiotic adjustments as either deescalation, escalation, or no change. Results The percentage of cases deescalated was higher in response to physician (house staff or attending physician) notification of pathogen susceptibility information when compared with a response to pathogen identification alone (33.2% vs 22.6%). Empiric antibiotics were not altered within 24 hours in response to pathogen identification in 70.7% of cases and were not altered within 24 hours in response to pathogen sensitivity determination in 58.6% of cases. However, when considering the time frame from when empiric antibiotics were started to 24 hours after notification of susceptibility information, 49.5% of cases were deescalated and 41.5% of cases had no net change in the antibiotic spectrum score. The magnitude of deescalations were notably larger than escalations. The mean (SD) time to deescalation of antibiotic coverage was shorter (P =.049) in response to pathogen identification at 8 (7.4) hours compared with sensitivity information at 10.4 (7) hours, but may not be clinically relevant. Conclusion Health care providers at the Minneapolis VAMC appear to be using positive blood culture results in a timely fashion consistent with best practices. Because empirically initiated antibiotics typically are broad in spectrum, the magnitude of deescalations were notably larger than escalations. Adherence to these standards may be a reflection of the infectious disease staff oversight of antibiotic administration. Furthermore, the systems outlined in this quality improvement study may be replicated at other VAMCs across the country by either in-house infectious disease staff or through remote monitoring of the electronic health record by other infectious disease experts at a more centralized VAMC. Widespread adoption throughout the Veterans Health Administration may result in improved antibiotic resistance profiles and better clinical outcomes for our nation's veterans.
Collapse
Affiliation(s)
- Shaun Heimbichner Johnson
- is a Medical Student at Case Western Reserve University School of Medicine in Cleveland, Ohio. is a Surgeon and Assistant Service Chief at the Minneapolis Veterans Affairs Health Care System in Minnesota and an Assistant Professor of Surgery at the University of Minnesota
| | - Steven James Waisbren
- is a Medical Student at Case Western Reserve University School of Medicine in Cleveland, Ohio. is a Surgeon and Assistant Service Chief at the Minneapolis Veterans Affairs Health Care System in Minnesota and an Assistant Professor of Surgery at the University of Minnesota
| |
Collapse
|
11
|
Haider A, Alavi F, Siddiqa A, Abbas H, Patel H. Fulminant Pseudomembranous Colitis Leading to Clostridium Paraputrificum Bacteremia. Cureus 2021; 13:e13763. [PMID: 33842139 PMCID: PMC8022762 DOI: 10.7759/cureus.13763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Clostridium species are spore-forming gram-positive anaerobic rod bacteria that cause a broad range of infections in humans, including intra-abdominal infections, myonecrosis, and bacteremia. Pseudomembranous colitis (PMC) is a severe form of infection caused by Clostridioides difficile. Clostridial bacteremia usually occurs in the settings of neutropenia, alcohol abuse, diabetes mellitus, sickle cell anemia, malignancy, hemodialysis, inflammatory bowel disease, and AIDS. We report a case of fulminant PMC leading to C. paraputrificum bacteremia in an otherwise immunocompetent patient. To our knowledge, this is the first case report of such an occurrence.
Collapse
Affiliation(s)
- Asim Haider
- Internal Medicine, BronxCare Health System, Bronx, USA
| | - Fareeha Alavi
- Internal Medicine, BronxCare Health System, Bronx, USA
| | | | - Hafsa Abbas
- Gastroenterology, BronxCare Health System, Bronx, USA
| | - Harish Patel
- Gastroenterology, BronxCare Health System, Bronx, USA
| |
Collapse
|
12
|
Gupta A, Ananthakrishnan AN. Economic burden and cost-effectiveness of therapies for Clostridiodes difficile infection: a narrative review. Therap Adv Gastroenterol 2021; 14:17562848211018654. [PMID: 34104214 PMCID: PMC8170348 DOI: 10.1177/17562848211018654] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/30/2021] [Indexed: 02/04/2023] Open
Abstract
Clostridioides difficile is the most common cause of healthcare-associated diarrhea. Disease complications as well as recurrent infections contribute significantly to morbidity and mortality. Over the past decades, there has been a rapid increase in the incidence of C. difficile infection (CDI), with a rise in the number of community-acquired cases. CDI has a profound economic impact on both the healthcare system and patients, secondary to recurrences, hospitalization, prolonged length of stay, cost of treatment, and indirect societal costs. With emergence of newer treatment options, the standard of care is shifting from metronidazole and vancomycin towards fidaxomicin and fecal microbiota transplantation (FMT), which despite being more expensive, are more efficacious in preventing recurrences and hence overall are more beneficial forms of therapy per cost-effectiveness analyses. Data regarding preferred route of FMT, timing of FMT, and non-conventional therapies such as bezlotoxumab is scant. There is a need for further studies to elucidate the true attributable costs of CDI as well as continued cost-effectiveness research to reduce the economic burden associated with the disease and improve clinical practice.
Collapse
Affiliation(s)
- Akshita Gupta
- Department of Medicine, Massachusetts General
Hospital, Boston, MA, USA
| | | |
Collapse
|
13
|
Lei C, Mu J, Teng Y, He L, Xu F, Zhang X, Sundaram K, Kumar A, Sriwastva MK, Lawrenz MB, Zhang L, Yan J, Feng W, McClain CJ, Zhang X, Zhang HG. Lemon Exosome-like Nanoparticles-Manipulated Probiotics Protect Mice from C. d iff Infection. iScience 2020; 23:101571. [PMID: 33083738 PMCID: PMC7530291 DOI: 10.1016/j.isci.2020.101571] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/13/2020] [Accepted: 09/13/2020] [Indexed: 01/09/2023] Open
Abstract
Clostridioides difficile (C. diff) is the leading cause of antibiotic-associated colitis. Here, we report that lemon exosome-like nanoparticles (LELNs) manipulated probiotics to inhibit C. diff infection (CDI). LELN-manipulated Lactobacillus rhamnosus GG (LGG) and Streptococcus thermophilus ST-21 (STH) (LELN-LS) decrease CDI mortality via an LELN-mediated increase in bile resistance and gut survivability. LELN-LS treatment increases the AhR ligands indole-3-lactic acid (I3LA) and indole-3-carboxaldehyde (I3Ald), leading to induction of IL-22, and increases lactic acid leading to a decrease of C. diff fecal shedding by inhibiting C. diff growth and indole biosynthesis. A synergistic effect between STH and LGG was identified. The STH metabolites inhibit gluconeogenesis of LGG and allow fructose-1,6-bisphosphate (FBP) to accumulate in LGG; accumulated FBP then activates lactate dehydrogenase of LGG (LGG-LDH) and enhances production of lactic acid and the AhR ligand. Our findings provide a new strategy for CDI prevention and treatment with a new type of prebiotics.
LELNs-manipulated probiotics protect mice from C. diff infection LELNs manipulation modulates gut metabolomics composition Cross talk between LGG and STH enhances production of lactic acid and AhR ligands
Collapse
Affiliation(s)
- Chao Lei
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Jingyao Mu
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Yun Teng
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Liqing He
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Fangyi Xu
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Xiangcheng Zhang
- Department of ICU, the Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu 223300, China
| | - Kumaran Sundaram
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Anil Kumar
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Mukesh K Sriwastva
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Matthew B Lawrenz
- Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, USA.,Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Lifeng Zhang
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Jun Yan
- Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA
| | - Wenke Feng
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Craig J McClain
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Xiang Zhang
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Huang-Ge Zhang
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40206, USA.,Department of Chemistry, University of Louisville, CTRB 309 505 Hancock Street, Louisville, KY 40202, USA.,Department of Microbiology & Immunology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
14
|
Evans CT, Fitzpatrick M, Ramanathan S, Kralovic SM, Burns SP, Goldstein B, Smith B, Gerding DN, Johnson S. Healthcare facility-onset, healthcare facility-associated Clostridioides difficile infection in Veterans with spinal cord injury and disorder. J Spinal Cord Med 2020; 43:642-652. [PMID: 31663843 PMCID: PMC7534364 DOI: 10.1080/10790268.2019.1672953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: To describe the burden and risk of healthcare facility-onset, healthcare facility-associated (HO-HCFA) Clostridioides difficile infection (CDI) in Veterans with spinal cord injury and disorder (SCI/D). Design: Retrospective, longitudinal cohort study from October 1, 2001-September 30, 2010. Setting: Ninety-four acute care Veterans Affairs facilities. Participants: Patients with SCI/D. Outcomes: Incidence rate of HO-HCFA CDI. Methods: Rates of CDI were determined, and crude unadjusted incidence rate ratios (IRRs) and 95% confidence intervals (CIs) were calculated. Multivariable Poisson random-effects regression analyses were used to assess factors independently associated with the rate of CDI. Results: 1,409 cases of HO-HCFA CDI were identified. CDI rates in 2002 were 13.9/10,000 person-days and decreased to 5.5/10,000 person-days by 2010. Multivariable regression analyses found that antibiotic (IRR = 18.79, 95% CI 14.09-25.07) and proton-pump inhibitor (PPI) or H2 blocker use (IRR = 7.71, 95% CI 5.47-10.86) were both independently associated with HO-HCFA CDI. Exposure to both medications demonstrated a synergistic risk (IRR = 37.55, 95% CI 28.39-49.67). Older age, Northeast region, and invasive respiratory procedure in the prior 30 days were also independent risk factors, while longer SCI duration and care at a SCI center were protective. Conclusion: Although decreasing, CDI rates in patients with SCI/D remain high. Targeted antimicrobial stewardship and pharmacy interventions that reduce antibiotic and PPI/H2 blocker use could have profound benefits in decreasing HO-HCFA CDI in this high-risk population.
Collapse
Affiliation(s)
- Charlesnika T. Evans
- Department of Veterans Affairs, Center of Innovation for Complex Chronic Healthcare, Edward Hines Jr VA Hospital, Hines, Illinois, USA
- Department of Preventive Medicine and Center for Healthcare Studies, Northwestern University, Chicago, Illinois, USA
| | - Margaret Fitzpatrick
- Department of Veterans Affairs, Center of Innovation for Complex Chronic Healthcare, Edward Hines Jr VA Hospital, Hines, Illinois, USA
- Department of Medicine, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Swetha Ramanathan
- Department of Veterans Affairs, Center of Innovation for Complex Chronic Healthcare, Edward Hines Jr VA Hospital, Hines, Illinois, USA
| | - Stephen M. Kralovic
- Cincinnati VA Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Stephen P. Burns
- VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA
| | | | - Bridget Smith
- Department of Veterans Affairs, Center of Innovation for Complex Chronic Healthcare, Edward Hines Jr VA Hospital, Hines, Illinois, USA
- Department of Pediatrics and Center for Community Health, Northwestern University, Chicago, Illinois, USA
| | - Dale N. Gerding
- Department of Veterans Affairs, Research Service, Edward Hines Jr VA Hospital, Hines, Illinois, USA
| | - Stuart Johnson
- Department of Medicine, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
- Department of Veterans Affairs, Research Service, Edward Hines Jr VA Hospital, Hines, Illinois, USA
| |
Collapse
|
15
|
Singla A, Pash D. Diarrhea or No Diarrhea, It Still Hurts: An Atypical Case of Clostridioides difficile. Cureus 2020; 12:e9900. [PMID: 32850262 PMCID: PMC7444969 DOI: 10.7759/cureus.9900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Clostridioides difficile (C. difficile) is a common nosocomial infection that is classically described as profuse watery diarrhea in hospitalized patients after antibiotic use. We present a case of a 76-year-old female who presented to our emergency room with diffuse abdominal pain after consuming meals. This patient had completed treatment with oral vancomycin for C. difficile infection two weeks prior to admission and had been asymptomatic until this point. After receiving treatments for presumed acute mesenteric ischemia did not yield clinical improvement, polymerase chain reaction for C. difficile stool antigen was tested and was positive. While the patient did not have diarrhea, the classical feature of C. difficile infection, she quickly improved after treatment with oral vancomycin.
Collapse
|
16
|
Chung H, Jung J, Kim MJ, Sung H, Kim MN, Chong YP, Kim SH, Lee SO, Kim YS, Woo JH, Choi SH. Clinical characteristics and prognostic factors of extraintestinal infection caused by Clostridioides difficile: analysis of 60 consecutive cases. Eur J Clin Microbiol Infect Dis 2020; 39:2133-2141. [PMID: 32632700 DOI: 10.1007/s10096-020-03975-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/30/2020] [Indexed: 01/03/2023]
Abstract
Data regarding extraintestinal Clostridioides difficile infections (ECDIs) remain scarce and anecdotal. We conducted a retrospective cohort study to investigate characteristics and prognostic factors in patients with ECDI. From January 1997 through December 2018, 60 patients were enrolled and divided into three groups as follows: group A (gastrointestinal [GI] disruption caused by malignancy, n = 13); group B (GI disruption from causes other than malignancy, n = 25); group C (no GI disruption, n = 22). GI disruption was defined as compromised integrity of the GI tract caused by abdominal surgery, perforation, malignancy, enterocolitis, or bleeding. The incidence of ECDI was 2.53 per 100,000 admissions. The most common specimens yielded C. difficile were blood (36.7%), peritoneal fluid (20.0%), and abscesses (16.7%). Six patients (10.0%) had confirmed C. difficile enterocolitis, and 36 patients (60.0%) had a polymicrobial infection. C. difficile bacteremia was significantly more common in group A patients than those in groups B or C (53.8% vs. 48.0% vs. 13.6%, p = 0.02), as was the 30-day mortality rate (69.2% vs. 12.0% vs. 18.2%, respectively; p < 0.001). In multivariate analysis, group A (adjusted odds ratio [aOR], 17.32; 95% confidence interval [CI], 2.96-101.21; p = 0.002) and an age of > 65 years (aOR, 7.09; 95% CI, 1.31-38.45; p = 0.02) were independent risk factors for 30-day mortality. ECDI was uncommonly associated with C. difficile enterocolitis. Two factors, GI disruption caused by malignancy, and old age, were associated with significantly poorer short-term outcomes.
Collapse
Affiliation(s)
- Hyemin Chung
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Jiwon Jung
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Min Jae Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Heungsup Sung
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi-Na Kim
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong Pil Chong
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Sang-Oh Lee
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Yang Soo Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Jun Hee Woo
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea
| | - Sang-Ho Choi
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Seoul, 05505, Republic of Korea.
| |
Collapse
|
17
|
Miller AC, Segre AM, Pemmeraju SV, Sewell DK, Polgreen PM. Association of Household Exposure to Primary Clostridioides difficile Infection With Secondary Infection in Family Members. JAMA Netw Open 2020; 3:e208925. [PMID: 32589232 PMCID: PMC7320299 DOI: 10.1001/jamanetworkopen.2020.8925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/14/2020] [Indexed: 12/14/2022] Open
Abstract
Importance Clostridioides difficile infection (CDI) is a common hospital-acquired infection. Whether family members are more likely to experience a CDI following CDI in another separate family member remains to be studied. Objective To determine the incidence of potential family transmission of CDI. Design, Setting, and Participants In this case-control study comparing the incidence of CDI among individuals with prior exposure to a family member with CDI to those without prior family exposure, individuals were binned into monthly enrollment strata based on exposure status (eg, family exposure) and confounding factors (eg, age, prior antibiotic use). Data were derived from population-based, longitudinal commercial insurance claims from the Truven Marketscan Commercial Claims and Encounters and Medicare Supplemental databases from 2001 to 2017. Households with at least 2 family members continuously enrolled for at least 1 month were eligible. CDI incidence was computed within each stratum. A regression model was used to compare incidence of CDI while controlling for possible confounding characteristics. Exposures Index CDI cases were identified using inpatient and outpatient diagnosis codes. Exposure risks 60 days prior to infection included CDI diagnosed in another family member, prior hospitalization, and antibiotic use. Main Outcomes and Measures The primary outcome was the incidence of CDI in a given monthly enrollment stratum. Separate analyses were considered for CDI diagnosed in outpatient or hospital settings. Results A total of 224 818 cases of CDI, representing 194 424 enrollees (55.9% female; mean [SD] age, 52.8 [22.2] years) occurred in families with at least 2 enrollees. Of these, 1074 CDI events (4.8%) occurred following CDI diagnosis in a separate family member. Prior family exposure was significantly associated with increased incidence of CDI, with an incidence rate ratio (IRR) of 12.47 (95% CI, 8.86-16.97); this prior family exposure represented the factor with the second highest IRR behind hospital exposure (IRR, 16.18 [95% CI, 15.31-17.10]). For community-onset CDI cases without prior hospitalization, the IRR for family exposure was 21.74 (95% CI, 15.12-30.01). Age (IRR, 9.90 [95% CI, 8.92-10.98] for ages ≥65 years compared with ages 0-17 years), antibiotic use (IRR, 3.73 [95% CI, 3.41-4.08] for low-risk and 14.26 [95% CI, 13.27-15.31] for high-risk antibiotics compared with no antibiotics), and female sex (IRR, 1.44 [95% CI, 1.36-1.53]) were also positively associated with incidence. Conclusions and Relevance This study found that individuals with family exposure may be at significantly greater risk for acquiring CDI, which highlights the importance of the shared environment in the transmission and acquisition of C difficile.
Collapse
Affiliation(s)
| | | | | | | | - Philip M. Polgreen
- Department of Epidemiology, University of Iowa, Iowa City
- Department of Internal Medicine, University of Iowa, Iowa City
| |
Collapse
|
18
|
Molecular Characterization and Diagnosis of Nosocomial Clostridium difficile Infection in Hospitalized Patients. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2020. [DOI: 10.5812/archcid.97330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Bouza E, Aguado JM, Alcalá L, Almirante B, Alonso-Fernández P, Borges M, Cobo J, Guardiola J, Horcajada JP, Maseda E, Mensa J, Merchante N, Muñoz P, Pérez Sáenz JL, Pujol M, Reigadas E, Salavert M, Barberán J. Recommendations for the diagnosis and treatment of Clostridioides difficile infection: An official clinical practice guideline of the Spanish Society of Chemotherapy (SEQ), Spanish Society of Internal Medicine (SEMI) and the working group of Postoperative Infection of the Spanish Society of Anesthesia and Reanimation (SEDAR). REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2020; 33:151-175. [PMID: 32080996 PMCID: PMC7111242 DOI: 10.37201/req/2065.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 01/26/2020] [Indexed: 12/12/2022]
Abstract
This document gathers the opinion of a multidisciplinary forum of experts on different aspects of the diagnosis and treatment of Clostridioides difficile infection (CDI) in Spain. It has been structured around a series of questions that the attendees considered relevant and in which a consensus opinion was reached. The main messages were as follows: CDI should be suspected in patients older than 2 years of age in the presence of diarrhea, paralytic ileus and unexplained leukocytosis, even in the absence of classical risk factors. With a few exceptions, a single stool sample is sufficient for diagnosis, which can be sent to the laboratory with or without transportation media for enteropathogenic bacteria. In the absence of diarrhoea, rectal swabs may be valid. The microbiology laboratory should include C. difficile among the pathogens routinely searched in patients with diarrhoea. Laboratory tests in different order and sequence schemes include GDH detection, presence of toxins, molecular tests and toxigenic culture. Immediate determination of sensitivity to drugs such as vancomycin, metronidazole or fidaxomycin is not required. The evolution of toxin persistence is not a suitable test for follow up. Laboratory diagnosis of CDI should be rapid and results reported and interpreted to clinicians immediately. In addition to the basic support of all diarrheic episodes, CDI treatment requires the suppression of antiperistaltic agents, proton pump inhibitors and antibiotics, where possible. Oral vancomycin and fidaxomycin are the antibacterials of choice in treatment, intravenous metronidazole being restricted for patients in whom the presence of the above drugs in the intestinal lumen cannot be assured. Fecal material transplantation is the treatment of choice for patients with multiple recurrences but uncertainties persist regarding its standardization and safety. Bezlotoxumab is a monoclonal antibody to C. difficile toxin B that should be administered to patients at high risk of recurrence. Surgery is becoming less and less necessary and prevention with vaccines is under research. Probiotics have so far not been shown to be therapeutically or preventively effective. The therapeutic strategy should be based, rather than on the number of episodes, on the severity of the episodes and on their potential to recur. Some data point to the efficacy of oral vancomycin prophylaxis in patients who reccur CDI when systemic antibiotics are required again.
Collapse
Affiliation(s)
- E Bouza
- Emilio Bouza MD, PhD, Instituto de Investigación Sanitaria Gregorio Marañón, Servicio de Microbiología Clínica y E. Infecciosas C/ Dr. Esquerdo, 46 - 28007 Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Baker SJ, Chu DI. Physical, Laboratory, Radiographic, and Endoscopic Workup for Clostridium difficile Colitis. Clin Colon Rectal Surg 2020; 33:82-86. [PMID: 32104160 DOI: 10.1055/s-0039-3400474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Clostridium (reclassified as " Clostridioides ") difficile colitis is a common nosocomial infection associated with increased morbidity and mortality. Like many clinical encounters, a focused history and physical examination will help to guide initial management. Further laboratory testing will assist with diagnosis through stool studies, and blood tests, such as white blood cell counts and serum creatinine, can help to stratify patients into illness severity groups for treatment decisions. Radiographic evaluation can be helpful in patients with severe disease and concern for complicated colitis. Endoscopic evaluation should be carefully considered in patients with suspected mucosal injury secondary to infections and plays a role when an alternative diagnosis is suspected. Treatment options depend on the clinical presentation and can range from antibiotic therapy to emergent surgery to fecal transplantation for recurrent episodes. Care for these patients is often challenging, but through a systemic workup the appropriate treatment may be delivered.
Collapse
Affiliation(s)
- Samantha J Baker
- Department of General Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel I Chu
- Department of Gastrointestinal Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
21
|
Lall S, Nataraj G, Mehta P. Use of culture- and ELISA-based toxin assay for detecting Clostridium Difficile, a neglected pathogen: A single-center study from a tertiary care setting. J Lab Physicians 2020; 9:254-259. [PMID: 28966486 PMCID: PMC5607753 DOI: 10.4103/jlp.jlp_157_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION: Clostridium difficile is a Gram-positive spore-bearing anaerobic bacillus increasingly associated with both community- and hospital-acquired colitis and diarrhea. It is the most common identifiable bacterial cause of healthcare-associated diarrhea associated with antibiotic use and one of the most common anaerobic infections. The diagnosis of C. difficile infection includes detection of toxin A/B in stool specimens by direct enzyme immunoassay, culture of pathogen from the stool specimens using a selective agar Cycloserine-Cefoxitin fructose agar (CCFA), tissue culture assay, and detection of glutamate dehydrogenase an enzyme produced by C. difficile. With few reports from India on this disease, the present study was planned to throw more light on the prevalence and utility of laboratory diagnostic methods for C. difficile-associated diarrhea (CDAD). MATERIAL AND METHODS: After taking approval from the Ethics Committee, 150 patients with antibiotic-associated diarrhea were taken as a study group and fifty patients with exposure to antibiotics but who did not develop diarrhea were taken as controls. Stool specimen was processed for both culture on CCFA and toxin detection by IVD Tox A + B ELISA. RESULTS: Only four specimens were culture positive, whereas 13 were ELISA positive. All culture-positive isolates were toxigenic. C. difficile was neither isolated nor its toxin detected in the control group. Culture- and toxin-based assays may not detect all cases of CDAD. CONCLUSION: Based on the results of the present study, culture does not provide any additional yield over toxin assay. Better diagnostic modalities would be required to prove CDAD.
Collapse
Affiliation(s)
- Sujata Lall
- Department of Microbiology, Seth GSMC and KEMH, Mumbai, Maharashtra, India
| | - Gita Nataraj
- Department of Microbiology, Seth GSMC and KEMH, Mumbai, Maharashtra, India
| | - Preeti Mehta
- Department of Microbiology, Seth GSMC and KEMH, Mumbai, Maharashtra, India
| |
Collapse
|
22
|
Kusachi S, Watanabe M, Asai K, Kiribayashi T, Niitsuma T, Nishimuta H, Saida Y. Successful perioperative infection control measures after gastroenterological surgery reduced the number of cases of methicillin-resistant Staphylococcus aureus or Clostridioides (Clostridium) difficile infection to almost zero over a 30-year period: a single-department experience. Surg Today 2019; 50:258-266. [PMID: 31642991 DOI: 10.1007/s00595-019-01899-2] [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: 05/07/2019] [Accepted: 08/11/2019] [Indexed: 11/27/2022]
Abstract
PURPOSE To investigate changes in the incidence of postoperative infections in the surgical department of a teaching hospital. METHODS During the 30-year period from September 1987 to August 2017, 11,568 gastroenterological surgical procedures were performed in our surgical department. This 30-year period was divided into seven periods (A-G), ranging from 2 to 7 years each and based on the infection control methods used in each period. We then compared the rates of incisional surgical site infection (SSI) and organ/space SSI; remote infection (RI) including respiratory tract infection (RTI), intravascular catheter-related infection, and urinary tract infection (UTI); and antibiotic-associated colitis caused by methicillin-resistant Staphylococcus aureus (MRSA) enteritis or Clostridioides (Clostridium) difficile-associated disease (CDAD) among the seven periods. RESULTS In periods B (September 1990-August 1997) and E (November 2004-July 2007), when a unique antibiotic therapy devised in our department was in use, MRSA was isolated from only 0.3% and 0.4% of surgical patients, respectively, and these rates were significantly lower than those in the other periods (p < 0.05). The rate of CDAD increased during period F (August 2007-July 2014), but in period G (August 2014-August 2017), restrictions were placed on the use of antibiotics with a strong anti-anaerobic action and, in this period, the rate of CDAD was only 0.04%, which was significantly lower than that in period F (p < 0.05). CONCLUSIONS Limiting the use of antibiotics that tend to disrupt the intestinal flora may reduce the rates of MRSA infection and CDAD after gastroenterological surgery.
Collapse
Affiliation(s)
- Shinya Kusachi
- Department of Surgery, Tohokamagaya Hospital, 594 Awano, Kamagaya-si, Chiba-Ken, 273-0132, Japan.
| | - Manabu Watanabe
- Department of Surgery, Toho University Medical Center Ohashi Hospital, Tokyo, Japan
| | - Koji Asai
- Department of Surgery, Toho University Medical Center Ohashi Hospital, Tokyo, Japan
| | - Takaharu Kiribayashi
- Department of Surgery, Toho University Medical Center Ohashi Hospital, Tokyo, Japan
| | - Toru Niitsuma
- Department of Surgery, Toho University Medical Center Ohashi Hospital, Tokyo, Japan
| | - Hironobu Nishimuta
- Department of Surgery, Toho University Medical Center Ohashi Hospital, Tokyo, Japan
| | - Yoshihisa Saida
- Department of Surgery, Toho University Medical Center Ohashi Hospital, Tokyo, Japan
| |
Collapse
|
23
|
Lal A, Davaro R, Mishra AK, Sahu KK, Abraham GM. Detection of coexisting toxigenic Clostridium difficile and nontyphoidal Salmonella in a healthcare worker with diarrhea: A therapeutic dilemma. J Family Med Prim Care 2019; 8:2724-2727. [PMID: 31548964 PMCID: PMC6753814 DOI: 10.4103/jfmpc.jfmpc_227_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 03/16/2019] [Accepted: 06/08/2019] [Indexed: 12/18/2022] Open
Abstract
Introduction: Clostridium difficile infection (CDI) and nontyphoidal Salmonella infection (NSI) have similar clinical manifestations and are seldom seen simultaneously. The decision-making in terms of antibiotic therapy becomes difficult when both the pathogens are isolated from the same patient. Case Presentation: We describe a case of Clostridium difficile (CD) enterocolitis in a healthcare provider who concomitantly tested positive for nontyphoidal Salmonella. Discussion: To the best of our knowledge after extensive literature review (English), this is only the fourth report highlighting this association. Conclusion: Although Salmonella is not a risk factor for CDI, it can cause intestinal inflammation and alteration in the intestinal flora. When two pathogens are isolated from the same patient, it is tempting to treat both with antibiotics as highlighted. When it involves healthcare workers, there is no difference in guidelines and should not be prescribed antibiotics with intent of reducing secondary transmission.
Collapse
Affiliation(s)
- Amos Lal
- Department of Internal Medicine, Saint Vincent Hospital, Worcester, MA, USA
| | - Raul Davaro
- Division of Infectious Diseases, Worcester Medical Center, Reliant Medical Group, Worcester, MA, USA
| | - Ajay Kumar Mishra
- Department of Internal Medicine, Saint Vincent Hospital, Worcester, MA, USA
| | - Kamal Kant Sahu
- Department of Medicine, Saint Vincent Hospital, Worcester, MA, USA
| | - George M Abraham
- Department of Medicine, Saint Vincent Hospital, Worcester, MA, USA
| |
Collapse
|
24
|
Mileto S, Das A, Lyras D. Enterotoxic Clostridia: Clostridioides difficile Infections. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0015-2018. [PMID: 31124432 PMCID: PMC11026080 DOI: 10.1128/microbiolspec.gpp3-0015-2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Indexed: 12/17/2022] Open
Abstract
Clostridioides difficile is a Gram-positive, anaerobic, spore forming pathogen of both humans and animals and is the most common identifiable infectious agent of nosocomial antibiotic-associated diarrhea. Infection can occur following the ingestion and germination of spores, often concurrently with a disruption to the gastrointestinal microbiota, with the resulting disease presenting as a spectrum, ranging from mild and self-limiting diarrhea to severe diarrhea that may progress to life-threating syndromes that include toxic megacolon and pseudomembranous colitis. Disease is induced through the activity of the C. difficile toxins TcdA and TcdB, both of which disrupt the Rho family of GTPases in host cells, causing cell rounding and death and leading to fluid loss and diarrhea. These toxins, despite their functional and structural similarity, do not contribute to disease equally. C. difficile infection (CDI) is made more complex by a high level of strain diversity and the emergence of epidemic strains, including ribotype 027-strains which induce more severe disease in patients. With the changing epidemiology of CDI, our understanding of C. difficile disease, diagnosis, and pathogenesis continues to evolve. This article provides an overview of the current diagnostic tests available for CDI, strain typing, the major toxins C. difficile produces and their mode of action, the host immune response to each toxin and during infection, animal models of disease, and the current treatment and prevention strategies for CDI.
Collapse
Affiliation(s)
- S Mileto
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| | - A Das
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| | - D Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia, 3800
| |
Collapse
|
25
|
Arato V, Gasperini G, Giusti F, Ferlenghi I, Scarselli M, Leuzzi R. Dual role of the colonization factor CD2831 in Clostridium difficile pathogenesis. Sci Rep 2019; 9:5554. [PMID: 30944377 PMCID: PMC6447587 DOI: 10.1038/s41598-019-42000-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/12/2019] [Indexed: 01/01/2023] Open
Abstract
Clostridium difficile is a Gram-positive, anaerobic bacterium and the leading cause of antibiotic-associated diarrhea and pseudomembranous colitis. C. difficile modulates its transition from a motile to a sessile lifestyle through a mechanism of riboswitches regulated by cyclic diguanosine monophosphate (c-di-GMP). Previously described as a sortase substrate positively regulated by c-di-GMP, CD2831 was predicted to be a collagen-binding protein and thus potentially involved in sessility. By overexpressing CD2831 in C. difficile and heterologously expressing it on the surface of Lactococcus lactis, here we further demonstrated that CD2831 is a collagen-binding protein, able to bind to immobilized collagen types I, III and V as well as native collagen produced by human fibroblasts. We also observed that the overexpression of CD2831 raises the ability to form biofilm on abiotic surface in both C. difficile and L. lactis. Notably, we showed that CD2831 binds to the collagen-like domain of the human complement component C1q, suggesting a role in preventing complement cascade activation via the classical pathway. This functional characterization places CD2831 in the Microbial Surface Components Recognizing Adhesive Matrix Molecule (MSCRAMMs) family, a class of virulence factors with a dual role in adhesion to collagen-rich tissues and in host immune evasion by binding to human complement components.
Collapse
Affiliation(s)
- Vanessa Arato
- Glaxo Smith Kline Vaccines, Via Fiorentina 1, 53100, Siena, Italy.,University of Padova, Department of Biomedical Sciences, 35131, Padua, Italy
| | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100, Siena, Italy
| | - Fabiola Giusti
- Glaxo Smith Kline Vaccines, Via Fiorentina 1, 53100, Siena, Italy
| | - Ilaria Ferlenghi
- Glaxo Smith Kline Vaccines, Via Fiorentina 1, 53100, Siena, Italy
| | - Maria Scarselli
- Glaxo Smith Kline Vaccines, Via Fiorentina 1, 53100, Siena, Italy
| | - Rosanna Leuzzi
- Glaxo Smith Kline Vaccines, Via Fiorentina 1, 53100, Siena, Italy.
| |
Collapse
|
26
|
Han S, Soylu MC, Kirimli CE, Wu W, Sen B, Joshi SG, Emery CL, Au G, Niu X, Hamilton R, Krevolin K, Shih WH, Shih WY. Rapid, label-free genetic detection of enteropathogens in stool without genetic isolation or amplification. Biosens Bioelectron 2019; 130:73-80. [PMID: 30731348 PMCID: PMC6469511 DOI: 10.1016/j.bios.2019.01.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/22/2018] [Accepted: 01/12/2019] [Indexed: 12/13/2022]
Abstract
Current genetic detection methods require gene isolation, gene amplification and detection with a fluorescent-tagged probe. They typically require sophisticated equipment and expensive fluorescent probes, rendering them not widely available for rapid acute infection diagnoses at the point of care to ensure timely treatment of the diseases. Here we report a rapid genetic detection method that can detect the bacterial gene directly from patient stools using a piezoelectric plate sensor (PEPS) in conjunction with a continuous flow system with two temperature zones. With stools spiked with sodium dodecyl sulfate (SDS) in situ bacteria lysing and DNA denaturation occurred in the high-temperature zone whereas in situ specific detection of the denatured DNA by the PEPS occurred in the lower-temperature zone. The outcome was a rapid genetic detection method that directly detected bacterial genes from stool in < 40 min without the need of gene isolation, gene amplification, or expensive fluorescent tag but with polymerase chain reaction (PCR) sensitivity. In 40 blinded patient stools, it detected the toxin B gene of Clostridium difficile with 95% sensitivity and 95% specificity. The all-electrical, label-free nature of the detection further supports its potential as a low-cost genetic test that can be used at the point of care.
Collapse
Affiliation(s)
- Song Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Mehmet C Soylu
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Ceyhun E Kirimli
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Wei Wu
- Department of Materials Science and Engineering, Drexel University, PA 19104, USA
| | - Bhaswati Sen
- Department of Microbiology and Immunology, Drexel University, Philadelphia, PA 10102, USA
| | - Suresh G Joshi
- Department of Microbiology and Immunology, Drexel University, Philadelphia, PA 10102, USA
| | | | - Giang Au
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Xiaomin Niu
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Richard Hamilton
- Department of Emergency Medicine, Drexel University, Philadelphia, PA 10102, USA
| | - Kyle Krevolin
- Microbiology & SIVM Laboratories, Hahnemann University Hospital, Philadelphia, PA 10102, USA
| | - Wei-Heng Shih
- Department of Materials Science and Engineering, Drexel University, PA 19104, USA
| | - Wan Y Shih
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Diorio C, Robinson PD, Ammann RA, Castagnola E, Erickson K, Esbenshade A, Fisher BT, Haeusler GM, Kuczynski S, Lehrnbecher T, Phillips R, Cabral S, Dupuis LL, Sung L. Guideline for the Management of Clostridium Difficile Infection in Children and Adolescents With Cancer and Pediatric Hematopoietic Stem-Cell Transplantation Recipients. J Clin Oncol 2018; 36:3162-3171. [PMID: 30216124 PMCID: PMC6209092 DOI: 10.1200/jco.18.00407] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The aim of this work was to develop a clinical practice guideline for the prevention and treatment of Clostridium difficile infection (CDI) in children and adolescents with cancer and pediatric hematopoietic stem-cell transplantation (HSCT) patients. METHODS An international multidisciplinary panel of experts in pediatric oncology and infectious diseases with patient advocate representation was convened. We performed systematic reviews of randomized controlled trials for the prevention or treatment of CDI in any population and considered the directness of the evidence to children with cancer and pediatric HSCT patients. We used the Grading of Recommendations Assessment, Development, and Evaluation approach to generate recommendations. RESULTS The panel made strong recommendations to administer either oral metronidazole or oral vancomycin for the initial treatment of nonsevere CDI and oral vancomycin for the initial treatment of severe CDI. Fidaxomicin may be considered in the setting of recurrent CDI. The panel suggested that probiotics not be routinely used for the prevention of CDI, and that monoclonal antibodies and probiotics not be routinely used for the treatment of CDI. A strong recommendation to not use fecal microbiota transplantation was made in this population. We identified key knowledge gaps and suggested directions for future research. CONCLUSION We present a guideline for the prevention and treatment of CDI in children and adolescents with cancer and pediatric HSCT patients. Future research should include randomized controlled trials that involve children with cancer and pediatric HSCT patients to improve the management of CDI in this population.
Collapse
Affiliation(s)
- Caroline Diorio
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Paula D. Robinson
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Roland A. Ammann
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Elio Castagnola
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Kelley Erickson
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Adam Esbenshade
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Brian T. Fisher
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Gabrielle M. Haeusler
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Susan Kuczynski
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Thomas Lehrnbecher
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Robert Phillips
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Sandra Cabral
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - L. Lee Dupuis
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom
| | - Lillian Sung
- Caroline Diorio, Paula D. Robinson, and Sandra Cabral, Pediatric Oncology Group of Ontario; Caroline Diorio, L. Lee Dupuis, and Lillian Sung, The Hospital for Sick Children; L. Lee Dupuis, University of Toronto, Toronto; Caroline Diorio, McMaster Children’s Hospital, Hamilton; Susan Kuczynski, Ontario Parents Advocating for Children with Cancer, Barrie, Ontario, Canada; Roland A. Ammann, Bern University Hospital, University of Bern, Bern, Switzerland; Elio Castagnola, Istituto Giannina Gaslini, Genova, Italy; Kelley Erickson and Brian T. Fisher, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA; Adam Esbenshade, Vanderbilt-Ingram Cancer Centre, Nashville, TN; Gabrielle M. Haeusler, Peter MacCallum Cancer Centre, Melbourne; Gabrielle M. Haeusler, Royal Children’s Hospital, Parkville; Gabrielle M. Haeusler, Paediatric Integrated Cancer Service, Victoria, Australia; Thomas Lehrnbecher, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany; Robert Phillips, Leeds Teaching Hospital, National Health Service Trust, Leeds; and Robert Phillips, University of York, York, United Kingdom.,Corresponding author: Lillian Sung, MD, PhD, Division of Haematology/Oncology, The Hospital for Sick Children, 555 University Ave, Toronto, ON M5G1X8, Canada; e-mail:
| |
Collapse
|
28
|
Muñoz-Miralles J, Trindade BC, Castro-Córdova P, Bergin IL, Kirk LA, Gil F, Aronoff DM, Paredes-Sabja D. Indomethacin increases severity of Clostridium difficile infection in mouse model. Future Microbiol 2018; 13:1271-1281. [PMID: 30238771 PMCID: PMC6190216 DOI: 10.2217/fmb-2017-0311] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/10/2018] [Indexed: 01/05/2023] Open
Abstract
AIM To evaluate the effect on the nonsteroidal anti-inflammatory drug indomethacin on Clostridium difficile infection (CDI) severity. MATERIALS & METHODS Indomethacin was administered in two different mouse models of antibiotic-associated CDI in two different facilities, using a low and high dose of indomethacin. RESULTS Indomethacin administration caused weight loss, increased the signs of severe infection and worsened histopathological damage, leading to 100% mortality during CDI. Indomethacin-treated, antibiotic-exposed mice infected with C. difficile had enhanced intestinal inflammation with increased expression of KC, IL-1β and IL-22 compared with infected mice unexposed to indomethacin. CONCLUSION These results demonstrate a negative impact of nonsteroidal anti-inflammatory drugs on antibiotic-associated CDI in mice and suggest that targeting the synthesis or signaling of prostaglandins might be an approach to ameliorating the severity of CDI.
Collapse
Affiliation(s)
- Juan Muñoz-Miralles
- Millennium Nucleus in the Biology of Intestinal Microbiota, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
- Microbiota-Host Interactions & Clostridia Research Group, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
| | - Bruno C Trindade
- Department of Pathology, The University of Massachusetts Medical School, Worcester, 01605 MA, USA
| | - Pablo Castro-Córdova
- Millennium Nucleus in the Biology of Intestinal Microbiota, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
- Microbiota-Host Interactions & Clostridia Research Group, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
| | - Ingrid L Bergin
- The Unit for Laboratory Animal Medicine, The University of Michigan, Ann Arbor, 48109 MI, USA
| | - Leslie A Kirk
- The Unit for Laboratory Animal Medicine, The University of Michigan, Ann Arbor, 48109 MI, USA
| | - Fernando Gil
- Millennium Nucleus in the Biology of Intestinal Microbiota, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
- Microbiota-Host Interactions & Clostridia Research Group, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
| | - David M Aronoff
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, 37232 TN, USA
| | - Daniel Paredes-Sabja
- Millennium Nucleus in the Biology of Intestinal Microbiota, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
- Microbiota-Host Interactions & Clostridia Research Group, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370186 Santiago, Chile
| |
Collapse
|
29
|
Clostridium difficile, the Difficult "Kloster" Fuelled by Antibiotics. Curr Microbiol 2018; 76:774-782. [PMID: 30084095 DOI: 10.1007/s00284-018-1543-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Clostridium difficile is normally present in low numbers in a healthy adult gastro-intestinal tract (GIT). Drastic changes in the microbial population, e.g., dysbiosis caused by extensive treatment with antibiotics, stimulates the growth of resistant strains and the onset of C. difficile infection (CDI). Symptoms of infection varies from mild diarrhea to colitis (associated with dehydration and bleeding), pseudomembranous colitis with yellow ulcerations in the mucosa of the colon, to fulminant colitis (perforation of the gut membrane), and multiple organ failure. Inflamed epithelial cells and damaged mucosal tissue predisposes the colon to other opportunistic pathogens such as Clostridium perfringens, Staphylococcus aureus, Klebsiella oxytoca, Candida spp., and Salmonella spp. This may lead to small intestinal bacterial overgrowth (SIBO), sepsis, toxic megacolon, and even colorectal cancer. Many stains of C. difficile are resistant to metronidazole and vancomycin. Vaccination may be an answer to CDI, but requires more research. Success in treatment with probiotics depends on the strains used. Oral or rectal fecal transplants are partly effective, as spores in the small intestine may germinate and colonize the colon. The effect of antibiotics on C. difficile and commensal gut microbiota is summarized and changes in gut physiology are discussed. The need to search for non-antibiotic methods in the treatment of CDI and C. difficile-associated disease (CDAD) is emphasized.
Collapse
|
30
|
Zhou P, Zhou N, Shao L, Li J, Liu S, Meng X, Duan J, Xiong X, Huang X, Chen Y, Fan X, Zheng Y, Ma S, Li C, Wu A. Diagnosis of Clostridium difficile infection using an UPLC-MS based metabolomics method. Metabolomics 2018; 14:102. [PMID: 30830376 DOI: 10.1007/s11306-018-1397-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 07/10/2018] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The fecal metabolome of Clostridium difficile (CD) infection is far from being understood, particularly its non-volatile organic compounds. The drawbacks of current tests used to diagnose CD infection hinder their application. OBJECTIVE The aims of this study were to find new characteristic fecal metabolites of CD infection and develop a metabolomics model for the diagnosis of CD infection. METHODS Ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) was used to characterize the fecal metabolome of CD positive and negative diarrhea and healthy control stool samples. RESULTS Diarrhea and healthy control samples showed distinct clusters in the principal components analysis score plot, and CD positive group and CD negative group demonstrated clearer separation in a partial least squares discriminate analysis model. The relative abundance of sphingosine, chenodeoxycholic acid, phenylalanine, lysophosphatidylcholine (C16:0), and propylene glycol stearate was higher, and the relative abundance of fatty amide, glycochenodeoxycholic acid, tyrosine, linoleyl carnitine, and sphingomyelin was lower in CD positive diarrhea groups, than in the CD negative group. A linear discriminant analysis model based on capsiamide, dihydrosphingosine, and glycochenodeoxycholic acid was further constructed to identify CD infection in diarrhea. The leave-one-out cross-validation accuracy and area under receiver operating characteristic curve for the training set/external validation set were 90.00/78.57%, and 0.900/0.7917 respectively. CONCLUSIONS Compared with other hospital-onset diarrhea, CD diarrhea has distinct fecal metabolome characteristics. Our UPLC-MS metabolomics model might be useful tool for diagnosing CD diarrhea.
Collapse
Affiliation(s)
- Pengcheng Zhou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Ning Zhou
- Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Li Shao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Jianzhou Li
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, People's Republic of China
| | - Sidi Liu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xiujuan Meng
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Juping Duan
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xinrui Xiong
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xun Huang
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Yuhua Chen
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Xuegong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yixiang Zheng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Shujuan Ma
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, 410078, Hunan, People's Republic of China
| | - Chunhui Li
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| | - Anhua Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| |
Collapse
|
31
|
Schäffler H, Breitrück A. Clostridium difficile - From Colonization to Infection. Front Microbiol 2018; 9:646. [PMID: 29692762 PMCID: PMC5902504 DOI: 10.3389/fmicb.2018.00646] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/19/2018] [Indexed: 12/19/2022] Open
Abstract
Clostridium difficile is the most frequent cause of nosocomial antibiotic-associated diarrhea. The incidence of C. difficile infection (CDI) has been rising worldwide with subsequent increases in morbidity, mortality, and health care costs. Asymptomatic colonization with C. difficile is common and a high prevalence has been found in specific cohorts, e.g., hospitalized patients, adults in nursing homes and in infants. However, the risk of infection with C. difficile differs significantly between these cohorts. While CDI is a clear indication for therapy, colonization with C. difficile is not believed to be a direct precursor for CDI and therefore does not require treatment. Antibiotic therapy causes alterations of the intestinal microbial composition, enabling C. difficile colonization and consecutive toxin production leading to disruption of the colonic epithelial cells. Clinical symptoms of CDI range from mild diarrhea to potentially life-threatening conditions like pseudomembranous colitis or toxic megacolon. While antibiotics are still the treatment of choice for CDI, new therapies have emerged in recent years such as antibodies against C. difficile toxin B and fecal microbial transfer (FMT). This specific therapy for CDI underscores the role of the indigenous bacterial composition in the prevention of the disease in healthy individuals and its role in the pathogenesis after alteration by antibiotic treatment. In addition to the pathogenesis of CDI, this review focuses on the colonization of C. difficile in the human gut and factors promoting CDI.
Collapse
Affiliation(s)
- Holger Schäffler
- Division of Gastroenterology, Department of Medicine II, University of Rostock, Rostock, Germany
| | - Anne Breitrück
- Extracorporeal Immunomodulation Unit, Fraunhofer Institute for Cell Therapy and Immunology, Rostock, Germany.,Institute of Medical Microbiology, Virology and Hygiene, University of Rostock, Rostock, Germany
| |
Collapse
|
32
|
Cotter KJ, Fan Y, Sieger GK, Weight CJ, Konety BR. Prevalence of Clostridium Difficile Infection in Patients After Radical Cystectomy and Neoadjuvant Chemotherapy. Bladder Cancer 2017; 3:305-310. [PMID: 29152554 PMCID: PMC5676759 DOI: 10.3233/blc-170132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background and Objectives: Clostridium Difficile is the most common cause of nosocomial infectious diarrhea. This study evaluates the prevalence and predictors of Clostridium Difficile infections in patients undergoing radical cystectomy with or without neoadjuvant chemotherapy. Methods: Retrospective chart review was performed of all patients undergoing cystectomy and urinary diversion at a single institution from 2011–2017. Infection was documented in all cases with testing for Clostridium Difficile polymerase chain reaction toxin B. Patient and disease related factors were compared for those who received neoadjuvant chemotherapy vs. those who did not in order to identify potential risk factors associated with C. Difficile infections. Chi squared test and logistic regression analysis were used to determine statistical significance. Results: Of 350 patients who underwent cystectomy, 41 (11.7%) developed Clostridium Difficile in the 30 day post-operative period. The prevalence of C. Difficile infection was higher amongst the patients undergoing cystectomy compared to the non-cystectomy admissions at our hospital (11.7 vs. 2.9%). Incidence was not significantly different among those who underwent cystectomy for bladder cancer versus those who underwent the procedure for other reasons. Median time to diagnosis was 6 days (range 3–28 days). The prevalence of C. Diff infections was not significantly different among those who received neoadjuvant chemotherapy vs. those who did not (11% vs. 10.4% p = 0.72). A significant association between C. Difficile infection was not seen with proton pump inhibitor use (p = 0.48), patient BMI (p = 0.67), chemotherapeutic regimen (p = 0.94), individual surgeon (p = 0.54), type of urinary diversion (0.41), or peri-operative antibiotic redosing (p = 0.26). Conclusions: Clostridium Difficile infection has a higher prevalence in patients undergoing cystectomy. No significant association between prevalence and exposure to neoadjuvant chemotherapy was seen.
Collapse
Affiliation(s)
| | - Yunhua Fan
- University of Minnesota Department of Urology, Minneapolis, MN, USA
| | | | | | | |
Collapse
|
33
|
Pizarro-Guajardo M, Díaz-González F, Álvarez-Lobos M, Paredes-Sabja D. Characterization of Chicken IgY Specific to Clostridium difficile R20291 Spores and the Effect of Oral Administration in Mouse Models of Initiation and Recurrent Disease. Front Cell Infect Microbiol 2017; 7:365. [PMID: 28856119 PMCID: PMC5557795 DOI: 10.3389/fcimb.2017.00365] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/28/2017] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile infection (CDI) are the leading cause of world-wide nosocomial acquired diarrhea. The current main clinical challenge in CDI is the elevated rate of infection recurrence that may reach up to 30% of the patients, which has been associated to the formation of dormant spores during the infection. We sought to characterize the effects of oral administration of specific anti-spore IgY in mouse models of CDI and recurrent CDI. The specificity of anti-spore IgY was evaluated in vitro. In both, initiation mouse model and recurrence mouse model, we evaluated the prophylactic and therapeutic effect of anti-spore IgY, respectively. Our results demonstrate that anti-spore IgY exhibited high specificity and titers against C. difficile spores and reduced spore adherence to intestinal cells in vitro. Administration of anti-spore IgY to C57BL/6 mice prior and during CDI delayed the appearance of the diarrhea by 1.5 day, and spore adherence to the colonic mucosa by 90%. Notably, in the recurrence model, co-administration of anti-spore IgY coupled with vancomycin delayed the appearance of recurrent diarrhea by a median of 2 days. Collectively, these observations suggest that anti-spore IgY antibodies may be used as a novel prophylactic treatment for CDI, or in combination with antibiotics to treat CDI and prevent recurrence of the infection.
Collapse
Affiliation(s)
- Marjorie Pizarro-Guajardo
- Microbiota-Host Interactions and Clostridia Research Group, Departamento de Ciencias Biologicas, Universidad Andres BelloSantiago, Chile
| | - Fernando Díaz-González
- Microbiota-Host Interactions and Clostridia Research Group, Departamento de Ciencias Biologicas, Universidad Andres BelloSantiago, Chile
| | - Manuel Álvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad CatólicaSantiago, Chile
| | - Daniel Paredes-Sabja
- Microbiota-Host Interactions and Clostridia Research Group, Departamento de Ciencias Biologicas, Universidad Andres BelloSantiago, Chile
| |
Collapse
|
34
|
Barker A, Ngam C, Musuuza J, Vaughn VM, Safdar N. Reducing Clostridium difficile in the Inpatient Setting: A Systematic Review of the Adherence to and Effectiveness of C. difficile Prevention Bundles. Infect Control Hosp Epidemiol 2017; 38:639-650. [PMID: 28343455 PMCID: PMC5654380 DOI: 10.1017/ice.2017.7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Clostridium difficile infection (CDI) is the most common infectious cause of nosocomial diarrhea, and its prevention is an urgent public health priority. However, reduction of CDI is challenging because of its complex pathogenesis, large reservoirs of colonized patients, and the persistence of infectious spores. The literature lacks high-quality evidence for evaluating interventions, and many hospitals have implemented bundled interventions to reduce CDI with variable results. Thus, we conducted a systematic review to examine the components of CDI bundles, their implementation processes, and their impact on CDI rates. METHODS We conducted a comprehensive literature search of multiple computerized databases from their date of inception through April 30, 2016. The protocol was registered in PROSPERO, an international prospective register of systematic reviews. Bundle effectiveness, adherence, and study quality were assessed for each study meeting our criteria for inclusion. RESULTS In the 26 studies that met the inclusion criteria for this review, implementation and adherence factors to interventions were variably and incompletely reported, making study reproducibility and replicability challenging. Despite contextual differences and the variety of bundle components utilized, all 26 studies reported an improvement in CDI rates. However, given the lack of randomized controlled trials in the literature, assessing a causal relationship between bundled interventions and CDI rates is currently impossible. CONCLUSION Cluster randomized trials that include a rigorous assessment of the implementation of bundled interventions are urgently needed to causally test the effect of intervention bundles on CDI rates. Infect Control Hosp Epidemiol 2017;38:639-650.
Collapse
Affiliation(s)
- Anna Barker
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Caitlyn Ngam
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jackson Musuuza
- Institute of Clinical and Translational Research, University of Wisconsin, Madison, WI, USA
| | - Valerie M. Vaughn
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- The Patient Safety Enhancement Program, University of Michigan and VA Ann Arbor Health System, Ann Arbor, MI, USA
| | - Nasia Safdar
- William S. Middleton Memorial Veterans Affairs Hospital, Madison, WI, USA
- Division of Infectious Diseases, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Infection Control, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| |
Collapse
|
35
|
Nelson RL, Suda KJ, Evans CT, Cochrane IBD Group. Antibiotic treatment for Clostridium difficile-associated diarrhoea in adults. Cochrane Database Syst Rev 2017; 3:CD004610. [PMID: 28257555 PMCID: PMC6464548 DOI: 10.1002/14651858.cd004610.pub5] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Clostridium difficile (C. difficile) is recognized as a frequent cause of antibiotic-associated diarrhoea and colitis. This review is an update of a previously published Cochrane review. OBJECTIVES The aim of this review is to investigate the efficacy and safety of antibiotic therapy for C. difficile-associated diarrhoea (CDAD), or C. difficile infection (CDI), being synonymous terms. SEARCH METHODS We searched MEDLINE, EMBASE, CENTRAL and the Cochrane IBD Group Specialized Trials Register from inception to 26 January 2017. We also searched clinicaltrials.gov and clinicaltrialsregister.eu for ongoing trials. SELECTION CRITERIA Only randomised controlled trials assessing antibiotic treatment for CDI were included in the review. DATA COLLECTION AND ANALYSIS Three authors independently assessed abstracts and full text articles for inclusion and extracted data. The risk of bias was independently rated by two authors. For dichotomous outcomes, we calculated the risk ratio (RR) and corresponding 95% confidence interval (95% CI). We pooled data using a fixed-effect model, except where significant heterogeneity was detected, at which time a random-effects model was used. The following outcomes were sought: sustained symptomatic cure (defined as initial symptomatic response and no recurrence of CDI), sustained bacteriologic cure, adverse reactions to the intervention, death and cost. MAIN RESULTS Twenty-two studies (3215 participants) were included. The majority of studies enrolled patients with mild to moderate CDI who could tolerate oral antibiotics. Sixteen of the included studies excluded patients with severe CDI and few patients with severe CDI were included in the other six studies. Twelve different antibiotics were investigated: vancomycin, metronidazole, fusidic acid, nitazoxanide, teicoplanin, rifampin, rifaximin, bacitracin, cadazolid, LFF517, surotomycin and fidaxomicin. Most of the studies were active comparator studies comparing vancomycin with other antibiotics. One small study compared vancomycin to placebo. There were no other studies that compared antibiotic treatment to a placebo or a 'no treatment' control group. The risk of bias was rated as high for 17 of 22 included studies. Vancomycin was found to be more effective than metronidazole for achieving symptomatic cure. Seventy-two per cent (318/444) of metronidazole patients achieved symptomatic cure compared to 79% (339/428) of vancomycin patients (RR 0.90, 95% CI 0.84 to 0.97; moderate quality evidence). Fidaxomicin was found to be more effective than vancomycin for achieving symptomatic cure. Seventy-one per cent (407/572) of fidaxomicin patients achieved symptomatic cure compared to 61% (361/592) of vancomycin patients (RR 1.17, 95% CI 1.04 to 1.31; moderate quality evidence). Teicoplanin may be more effective than vancomycin for achieving a symptomatic cure. Eightly-seven per cent (48/55) of teicoplanin patients achieved symptomatic cure compared to 73% (40/55) of vancomycin patients (RR 1.21, 95% CI 1.00 to 1.46; very low quality evidence). For other comparisons including the one placebo-controlled study the quality of evidence was low or very low due to imprecision and in many cases high risk of bias because of attrition and lack of blinding. One hundred and forty deaths were reported in the studies, all of which were attributed by study authors to the co-morbidities of the participants that lead to acquiring CDI. Although many other adverse events were reported during therapy, these were attributed to the participants' co-morbidities. The only adverse events directly attributed to study medication were rare nausea and transient elevation of liver enzymes. Recent cost data (July 2016) for a 10 day course of treatment shows that metronidazole 500 mg is the least expensive antibiotic with a cost of USD 13 (Health Warehouse). Vancomycin 125 mg costs USD 1779 (Walgreens for 56 tablets) compared to fidaxomicin 200 mg at USD 3453.83 or more (Optimer Pharmaceuticals) and teicoplanin at approximately USD 83.67 (GBP 71.40, British National Formulary). AUTHORS' CONCLUSIONS No firm conclusions can be drawn regarding the efficacy of antibiotic treatment in severe CDI as most studies excluded patients with severe disease. The lack of any 'no treatment' control studies does not allow for any conclusions regarding the need for antibiotic treatment in patients with mild CDI beyond withdrawal of the initiating antibiotic. Nonetheless, moderate quality evidence suggests that vancomycin is superior to metronidazole and fidaxomicin is superior to vancomycin. The differences in effectiveness between these antibiotics were not too large and the advantage of metronidazole is its far lower cost compared to the other two antibiotics. The quality of evidence for teicoplanin is very low. Adequately powered studies are needed to determine if teicoplanin performs as well as the other antibiotics. A trial comparing the two cheapest antibiotics, metronidazole and teicoplanin, would be of interest.
Collapse
Affiliation(s)
- Richard L Nelson
- University of Illinois School of Public HealthEpidemiology/Biometry Division1603 West TaylorRoom 956ChicagoIllinoisUSA60612
| | | | - Charlesnika T Evans
- Northwestern UniversityDepartment of Preventive Medicine and Center for Healthcare Studies633 N. St. ClairChicagoILUSA60611
| | | |
Collapse
|
36
|
Evolutionary clade affects resistance of Clostridium difficile spores to Cold Atmospheric Plasma. Sci Rep 2017; 7:41814. [PMID: 28155914 PMCID: PMC5290531 DOI: 10.1038/srep41814] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/30/2016] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile is a spore forming bacterium and the leading cause of colitis and antibiotic associated diarrhoea in the developed world. Spores produced by C. difficile are robust and can remain viable for months, leading to prolonged healthcare-associated outbreaks with high mortality. Exposure of C. difficile spores to a novel, non-thermal atmospheric pressure gas plasma was assessed. Factors affecting sporicidal efficacy, including percentage of oxygen in the helium carrier gas admixture, and the effect on spores from different strains representing the five evolutionary C. difficile clades was investigated. Strains from different clades displayed varying resistance to cold plasma. Strain R20291, representing the globally epidemic ribotype 027 type, was the most resistant. However all tested strains displayed a ~3 log reduction in viable spore counts after plasma treatment for 5 minutes. Inactivation of a ribotype 078 strain, the most prevalent clinical type seen in Northern Ireland, was further assessed with respect to surface decontamination, pH, and hydrogen peroxide concentration. Environmental factors affected plasma activity, with dry spores without the presence of organic matter being most susceptible. This study demonstrates that cold atmospheric plasma can effectively inactivate C. difficile spores, and highlights factors that can affect sporicidal activity.
Collapse
|
37
|
Kuntz JL, Smith DH, Petrik AF, Yang X, Thorp ML, Barton T, Barton K, Labreche M, Spindel SJ, Johnson ES. Predicting the Risk of Clostridium difficile Infection upon Admission: A Score to Identify Patients for Antimicrobial Stewardship Efforts. Perm J 2016; 20:20-5. [PMID: 26845084 DOI: 10.7812/tpp/15-049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Increasing morbidity and health care costs related to Clostridium difficile infection (CDI) have heightened interest in methods to identify patients who would most benefit from interventions to mitigate the likelihood of CDI. OBJECTIVE To develop a risk score that can be calculated upon hospital admission and used by antimicrobial stewards, including pharmacists and clinicians, to identify patients at risk for CDI who would benefit from enhanced antibiotic review and patient education. METHODS We assembled a cohort of Kaiser Permanente Northwest patients with a hospital admission from July 1, 2005, through December 30, 2012, and identified CDI in the six months following hospital admission. Using Cox regression, we constructed a score to identify patients at high risk for CDI on the basis of preadmission characteristics. We calculated and plotted the observed six-month CDI risk for each decile of predicted risk. RESULTS We identified 721 CDIs following 54,186 hospital admissions-a 6-month incidence of 13.3 CDIs/1000 patient admissions. Patients with the highest predicted risk of CDI had an observed incidence of 53 CDIs/1000 patient admissions. The score differentiated between patients who do and do not develop CDI, with values for the extended C-statistic of 0.75. Predicted risk for CDI agreed closely with observed risk. CONCLUSION Our risk score accurately predicted six-month risk for CDI using preadmission characteristics. Accurate predictions among the highest-risk patient subgroups allow for the identification of patients who could be targeted for and who would likely benefit from review of inpatient antibiotic use or enhanced educational efforts at the time of discharge planning.
Collapse
Affiliation(s)
| | - David H Smith
- Senior Investigator at The Center for Health Research in Portland, OR.
| | - Amanda F Petrik
- Research Associate at The Center for Health Research in Portland, OR.
| | - Xiuhai Yang
- Research Analyst at The Center for Health Research in Portland, OR.
| | - Micah L Thorp
- Nephrologist for Northwest Permanente in Portland, OR.
| | - Tracy Barton
- Infectious Disease Pharmacist at the Sunnyside Medical Center in Clackamas, OR.
| | - Karen Barton
- Infectious Disease Pharmacist at the Sunnyside Medical Center in Clackamas, OR.
| | - Matthew Labreche
- Infectious Disease Pharmacist at the Sunnyside Medical Center in Clackamas, OR.
| | - Steven J Spindel
- Infectious Disease Specialist at the Sunnyside Medical Center in Clackamas, OR.
| | - Eric S Johnson
- Investigator at The Center for Health Research in Portland, OR.
| |
Collapse
|
38
|
Schwarz S, Shen J, Kadlec K, Wang Y, Brenner Michael G, Feßler AT, Vester B. Lincosamides, Streptogramins, Phenicols, and Pleuromutilins: Mode of Action and Mechanisms of Resistance. Cold Spring Harb Perspect Med 2016; 6:a027037. [PMID: 27549310 PMCID: PMC5088508 DOI: 10.1101/cshperspect.a027037] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lincosamides, streptogramins, phenicols, and pleuromutilins (LSPPs) represent four structurally different classes of antimicrobial agents that inhibit bacterial protein synthesis by binding to particular sites on the 50S ribosomal subunit of the ribosomes. Members of all four classes are used for different purposes in human and veterinary medicine in various countries worldwide. Bacteria have developed ways and means to escape the inhibitory effects of LSPP antimicrobial agents by enzymatic inactivation, active export, or modification of the target sites of the agents. This review provides a comprehensive overview of the mode of action of LSPP antimicrobial agents as well as of the mutations and resistance genes known to confer resistance to these agents in various bacteria of human and animal origin.
Collapse
Affiliation(s)
- Stefan Schwarz
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), 31535 Neustadt-Mariensee, Germany
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| | - Jianzhong Shen
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| | - Kristina Kadlec
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), 31535 Neustadt-Mariensee, Germany
| | - Yang Wang
- Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing, P.R. China
| | - Geovana Brenner Michael
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), 31535 Neustadt-Mariensee, Germany
| | - Andrea T Feßler
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), 31535 Neustadt-Mariensee, Germany
| | - Birte Vester
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| |
Collapse
|
39
|
Minami K, Sakaguchi Y, Yoshida D, Yamamoto M, Ikebe M, Morita M, Toh Y. Successful treatments with polymyxin B hemoperfusion and recombinant human thrombomodulin for fulminant Clostridium difficile-associated colitis with septic shock and disseminated intravascular coagulation: a case report. Surg Case Rep 2016; 2:76. [PMID: 27468959 PMCID: PMC4965360 DOI: 10.1186/s40792-016-0199-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/01/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Clostridium difficile (CD)-associated colitis (CDAC) is endemic and a common nosocomial enteric disease encountered by surgeons in modern hospitals due to prophylactic or therapeutic antibiotic therapies. Currently, the incidence of fulminant CDAC, which readily causes septic shock followed by multiple organ dysfunction syndromes, is increasing. Fulminant CDAC requires surgeons to perform a prompt surgery, such as subtotal colectomy, to remove the septic source. It is known that fulminant CDAC is caused by the shift from an inflammatory response at a local mucosal level to a general systemic inflammatory reaction in which CD toxin-induced mediators' cascades disseminate. Recently, it has been proven that polymyxin B hemoperfusion (PMX-HP) improves septic shock and recombinant human thrombomodulin (rhTM) controls disseminated intravascular coagulation (DIC). In addition, clinically and basically, it has been shown that these treatments can control serous chemical mediators. Therefore, it is considered that these treatments are promising ones for patients with fulminant CDAC. In the current report, we present that these treatments without surgery contributed to the improvement of sepsis due to fulminant CDAC. CASE PRESENTATION We encountered a case who developed fulminant CDAC with septic shock and DIC after laparoscopic gastrectomy for gastric cancer. At admission to the intensive care unit, his APACHE II score was 22, which indicated an estimated risk of hospital death of 42.4 %. Our therapies were not the subtotal colectomy to remove septic sources but the combination treatments with both PMX-HP and rhTM. These combination therapies resulted in excellent outcomes, namely the dramatic improvement of septic shock and DIC and the patient's survival. We speculate that these combination therapies completely inhibit the CD toxin-induced mediators' cascades and correspond to the removal of septic sources. CONCLUSIONS We recommend both PMX-HP and rhTM for patients who develop fulminant CDAC with septic shock and DIC to increase the survival benefit and replace the need for surgical treatment.
Collapse
Affiliation(s)
- Kazuhito Minami
- Department of Gastroenterological Surgery, National Kyushu Cancer Center, Notame 3-1-1, Minami-ku, Fukuoka, 811-1395, Japan.
| | - Yoshihisa Sakaguchi
- Department of Gastroenterological Surgery, National Hospital Organization Kyushu Medical Center, Jigyohama 1-8-1, Chuo-ku, Fukuoka, 810-8563, Japan
| | - Daisuke Yoshida
- Department of Gastroenterological Surgery, National Kyushu Cancer Center, Notame 3-1-1, Minami-ku, Fukuoka, 811-1395, Japan
| | - Manabu Yamamoto
- Department of Gastroenterological Surgery, National Kyushu Cancer Center, Notame 3-1-1, Minami-ku, Fukuoka, 811-1395, Japan
| | - Masahiko Ikebe
- Department of Gastroenterological Surgery, National Kyushu Cancer Center, Notame 3-1-1, Minami-ku, Fukuoka, 811-1395, Japan
| | - Masaru Morita
- Department of Gastroenterological Surgery, National Kyushu Cancer Center, Notame 3-1-1, Minami-ku, Fukuoka, 811-1395, Japan
| | - Yasushi Toh
- Department of Gastroenterological Surgery, National Kyushu Cancer Center, Notame 3-1-1, Minami-ku, Fukuoka, 811-1395, Japan
| |
Collapse
|
40
|
Qiu H, Cassan R, Johnstone D, Han X, Joyee AG, McQuoid M, Masi A, Merluza J, Hrehorak B, Reid R, Kennedy K, Tighe B, Rak C, Leonhardt M, Dupas B, Saward L, Berry JD, Nykiforuk CL. Novel Clostridium difficile Anti-Toxin (TcdA and TcdB) Humanized Monoclonal Antibodies Demonstrate In Vitro Neutralization across a Broad Spectrum of Clinical Strains and In Vivo Potency in a Hamster Spore Challenge Model. PLoS One 2016; 11:e0157970. [PMID: 27336843 PMCID: PMC4919053 DOI: 10.1371/journal.pone.0157970] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/08/2016] [Indexed: 01/07/2023] Open
Abstract
Clostridium difficile (C. difficile) infection (CDI) is the main cause of nosocomial antibiotic-associated colitis and increased incidence of community-associated diarrhea in industrialized countries. At present, the primary treatment of CDI is antibiotic administration, which is effective but often associated with recurrence, especially in the elderly. Pathogenic strains produce enterotoxin, toxin A (TcdA), and cytotoxin, toxin B (TcdB), which are necessary for C. difficile induced diarrhea and gut pathological changes. Administration of anti-toxin antibodies provides an alternative approach to treat CDI, and has shown promising results in preclinical and clinical studies. In the current study, several humanized anti-TcdA and anti-TcdB monoclonal antibodies were generated and their protective potency was characterized in a hamster infection model. The humanized anti-TcdA (CANmAbA4) and anti-TcdB (CANmAbB4 and CANmAbB1) antibodies showed broad spectrum in vitro neutralization of toxins from clinical strains and neutralization in a mouse toxin challenge model. Moreover, co-administration of humanized antibodies (CANmAbA4 and CANmAbB4 cocktail) provided a high level of protection in a dose dependent manner (85% versus 57% survival at day 22 for 50 mg/kg and 20 mg/kg doses, respectively) in a hamster gastrointestinal infection (GI) model. This study describes the protective effects conferred by novel neutralizing anti-toxin monoclonal antibodies against C. difficile toxins and their potential as therapeutic agents in treating CDI.
Collapse
Affiliation(s)
- Hongyu Qiu
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Robyn Cassan
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Darrell Johnstone
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Xiaobing Han
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Antony George Joyee
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Monica McQuoid
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Andrea Masi
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - John Merluza
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Bryce Hrehorak
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Ross Reid
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Kieron Kennedy
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Bonnie Tighe
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Carla Rak
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Melanie Leonhardt
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Brian Dupas
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Laura Saward
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Jody D. Berry
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| | - Cory L. Nykiforuk
- Cangene Corporation, a subsidiary of Emergent BioSolutions Inc., 155 Innovation Drive, Winnipeg, MB, R3T 5Y3, Canada
| |
Collapse
|
41
|
Shin JH, Chaves-Olarte E, Warren CA. Clostridium difficile Infection. Microbiol Spectr 2016; 4:10.1128/microbiolspec.EI10-0007-2015. [PMID: 27337475 PMCID: PMC8118380 DOI: 10.1128/microbiolspec.ei10-0007-2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Indexed: 02/07/2023] Open
Abstract
Clostridium difficile is an anaerobic, Gram-positive, spore-forming, toxin-secreting bacillus that has long been recognized to be the most common etiologic pathogen of antibiotic-associated diarrhea. C. difficile infection (CDI) is now the most common cause of health care-associated infections in the United States and accounts for 12% of these infections (Magill SS et al., N Engl J Med370:1198-1208, 2014). Among emerging pathogens of public health importance in the United States, CDI has the highest population-based incidence, estimated at 147 per 100,000 (Lessa FC et al., N Engl J Med372:825-834, 2015). In a report on antimicrobial resistance, C. difficile has been categorized by the Centers for Disease Control and Prevention as one of three "urgent" threats (http://www.cdc.gov/drugresistance/threat-report-2013/). Although C. difficile was first described in the late 1970s, the past decade has seen the emergence of hypertoxigenic strains that have caused increased morbidity and mortality worldwide. Pathogenic strains, host susceptibility, and other regional factors vary and may influence the clinical manifestation and approach to intervention. In this article, we describe the global epidemiology of CDI featuring the different strains in circulation outside of North America and Europe where strain NAP1/027/BI/III had originally gained prominence. The elderly population in health care settings has been disproportionately affected, but emergence of CDI in children and healthy young adults in community settings has, likewise, been reported. New approaches in management, including fecal microbiota transplantation, are discussed.
Collapse
Affiliation(s)
- Jae Hyun Shin
- Department of Medicine, Division of Infectious Disease and International Health, University of Virginia, Charlottesville, VA 22908
| | - Esteban Chaves-Olarte
- Centro de Investigación en Enfermedades Tropicales, Facultad de Microbiología, Universidad de Costa Rica, Costa Rica
| | - Cirle A Warren
- Department of Medicine, Division of Infectious Disease and International Health, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
42
|
Zhu Z, Shi L, Feng H, Zhou HS. Single domain antibody coated gold nanoparticles as enhancer for Clostridium difficile toxin detection by electrochemical impedance immunosensors. Bioelectrochemistry 2016; 101:153-8. [PMID: 25460611 DOI: 10.1016/j.bioelechem.2014.10.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 10/06/2014] [Accepted: 10/18/2014] [Indexed: 12/19/2022]
Abstract
This work presents a sandwich-type electrochemical impedance immunosensor for detecting Clostridium difficile toxin A (TcdA) and toxin B (TcdB). Single domain antibody conjugated gold nanoparticles were applied to amplify the detection signal. Gold nanoparticles (Au NPs) were characterized by transmission electron microscopy and UV–vis spectra. The electron transfer resistance (Ret) of the working electrode surface was used as a parameter in the measurement of the biosensor. With the increase of the concentration of toxins from 1 pg/mL to 100 pg/mL, a linear relationship was observed between the relative electron transfer resistance and toxin concentration. In addition, the detection signal was enhanced due to the amplification effect. The limit of detection for TcdA and TcdB was found to be 0.61 pg/mL and 0.60 pg/mL respectively at a signal-to-noise ratio of 3 (S/N = 3). This method is simple, fast and ultrasensitive, thus possesses a great potential for clinical applications in the future.
Collapse
|
43
|
Abstract
Clostridium difficile is being recognized as a growing threat to many health-care systems. Epidemiology data shows that infection rates are soaring and the disease burden is increasing. Despite the efficacy of standard treatments, it is becoming evident that novel therapeutics will be required to tackle this disease. These new treatments aim to enhance the intestinal microbial barrier, activate the immune system and neutralize the toxins that mediate this disease. Many of these therapies are still in the beginning stages of investigation, however, in the next few years, more clinical data will become available to help implement many of these exciting new therapeutic approaches.
Collapse
Affiliation(s)
- David Padua
- a Department of Medicine , University of California, Los Angeles , Los Angeles , CA , USA
| | - Charalabos Pothoulakis
- a Department of Medicine , University of California, Los Angeles , Los Angeles , CA , USA
| |
Collapse
|
44
|
Poli A, Di Matteo S, Bruno GM, Fornai E, Valentino MC, Colombo GL. Economic burden of Clostridium difficile in five hospitals of the Florence health care system in Italy. Risk Manag Healthc Policy 2015; 8:207-13. [PMID: 26604846 PMCID: PMC4655953 DOI: 10.2147/rmhp.s90513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Despite the awareness about the increasing rates of Clostridium difficile infection (CDI) and the economic burden arising from its management (prolonged hospitalization, laboratory tests, visits, surgical treatment, environmental sanitation), few studies are available in Italy on the economic costs directly attributable to the CDI. The Florence health care system has designed a study with the aim of describing the costs attributable to the CDI and defines the incremental economic burden associated with the management of this complication. METHODS We conducted a retrospective study in five hospitals of the Florence health care system. The enrolled population included all patients who were hospitalized during the year 2013 with a diagnosis of CDI. Of the 187 total cases reported in 2013, 69 patients were enrolled, for whom the main cause of hospitalization was directly attributable to CDI. RESULTS We enrolled 69 patients (19 males and 50 females), with a mean age of 82.16 years (minimum 46 to maximum 98). The total number of hospitalization days observed was 886 (12.8 per patient on average). The data from this study show that the mean total incremental cost for a patient with CDI was €3,270.52 per year. The hospital stay length is the most significant cost parameter, having the largest influence on the overall costs, with an impact of 87% on the total cost. The results confirm the costs for the management of CDI in five hospitals of the Florence health care system are in line with data from the international literature. CONCLUSION The economic impact of CDI is most evident in the extension of the duration of hospitalization and emergency recurrences requiring new therapeutic options with the need to develop and implement new diagnostic and therapeutic algorithms in clinical practice.
Collapse
Affiliation(s)
- Anna Poli
- Vigilanza e Controllo Infezioni Correlate all'Assistenza, Ospedale Piero Palagi, Azienda Sanitaria di Firenze, Firenze, Italy
| | - Sergio Di Matteo
- SAVE Studi - Health Economics and Outcomes Research, Milan, Italy
| | - Giacomo M Bruno
- SAVE Studi - Health Economics and Outcomes Research, Milan, Italy
| | - Enrica Fornai
- Vigilanza e Controllo Infezioni Correlate all'Assistenza, Ospedale Piero Palagi, Azienda Sanitaria di Firenze, Firenze, Italy
| | | | - Giorgio L Colombo
- SAVE Studi - Health Economics and Outcomes Research, Milan, Italy ; Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
45
|
Furuya-Kanamori L, Marquess J, Yakob L, Riley TV, Paterson DL, Foster NF, Huber CA, Clements ACA. Asymptomatic Clostridium difficile colonization: epidemiology and clinical implications. BMC Infect Dis 2015; 15:516. [PMID: 26573915 PMCID: PMC4647607 DOI: 10.1186/s12879-015-1258-4] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 10/31/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The epidemiology of Clostridium difficile infection (CDI) has changed over the past decades with the emergence of highly virulent strains. The role of asymptomatic C. difficile colonization as part of the clinical spectrum of CDI is complex because many risk factors are common to both disease and asymptomatic states. In this article, we review the role of asymptomatic C. difficile colonization in the progression to symptomatic CDI, describe the epidemiology of asymptomatic C. difficile colonization, assess the effectiveness of screening and intensive infection control practices for patients at risk of asymptomatic C. difficile colonization, and discuss the implications for clinical practice. METHODS A narrative review was performed in PubMed for articles published from January 1980 to February 2015 using search terms 'Clostridium difficile' and 'colonization' or 'colonisation' or 'carriage'. RESULTS There is no clear definition for asymptomatic CDI and the terms carriage and colonization are often used interchangeably. The prevalence of asymptomatic C. difficile colonization varies depending on a number of host, pathogen, and environmental factors; current estimates of asymptomatic colonization may be underestimated as stool culture is not practical in a clinical setting. CONCLUSIONS Asymptomatic C. difficile colonization presents challenging concepts in the overall picture of this disease and its management. Individuals who are colonized by the organism may acquire protection from progression to disease, however they also have the potential to contribute to transmission in healthcare settings.
Collapse
Affiliation(s)
- Luis Furuya-Kanamori
- Research School of Population Health, The Australian National University, Building 62 Mills Road, Canberra, ACT 2601, Australia.
| | - John Marquess
- School of Population Health, The University of Queensland, Herston, QLD, Australia.
- Queensland Department of Health, Communicable Diseases Unit, Herston, QLD, Australia.
| | - Laith Yakob
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK.
| | - Thomas V Riley
- Microbiology and Immunology, School of Pathology and Laboratory Medicine, The University of Western Australia, Nedlands, WA, Australia.
- PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Nedlands, WA, Australia.
| | - David L Paterson
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.
| | - Niki F Foster
- PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Nedlands, WA, Australia.
| | - Charlotte A Huber
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.
| | - Archie C A Clements
- Research School of Population Health, The Australian National University, Building 62 Mills Road, Canberra, ACT 2601, Australia.
| |
Collapse
|
46
|
Polage CR, Gyorke CE, Kennedy MA, Leslie JL, Chin DL, Wang S, Nguyen HH, Huang B, Tang YW, Lee LW, Kim K, Taylor S, Romano PS, Panacek EA, Goodell PB, Solnick JV, Cohen SH. Overdiagnosis of Clostridium difficile Infection in the Molecular Test Era. JAMA Intern Med 2015; 175:1792-801. [PMID: 26348734 PMCID: PMC4948649 DOI: 10.1001/jamainternmed.2015.4114] [Citation(s) in RCA: 443] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE Clostridium difficile is a major cause of health care-associated infection, but disagreement between diagnostic tests is an ongoing barrier to clinical decision making and public health reporting. Molecular tests are increasingly used to diagnose C difficile infection (CDI), but many molecular test-positive patients lack toxins that historically defined disease, making it unclear if they need treatment. OBJECTIVE To determine the natural history and need for treatment of patients who are toxin immunoassay negative and polymerase chain reaction (PCR) positive (Tox-/PCR+) for CDI. DESIGN, SETTING, AND PARTICIPANTS Prospective observational cohort study at a single academic medical center among 1416 hospitalized adults tested for C difficile toxins 72 hours or longer after admission between December 1, 2010, and October 20, 2012. The analysis was conducted in stages with revisions from April 27, 2013, to January 13, 2015. MAIN OUTCOMES AND MEASURES Patients undergoing C difficile testing were grouped by US Food and Drug Administration-approved toxin and PCR tests as Tox+/PCR+, Tox-/PCR+, or Tox-/PCR-. Toxin results were reported clinically. Polymerase chain reaction results were not reported. The main study outcomes were duration of diarrhea during up to 14 days of treatment, rate of CDI-related complications (ie, colectomy, megacolon, or intensive care unit care) and CDI-related death within 30 days. RESULTS Twenty-one percent (293 of 1416) of hospitalized adults tested for C difficile were positive by PCR, but 44.7% (131 of 293) had toxins detected by the clinical toxin test. At baseline, Tox-/PCR+ patients had lower C difficile bacterial load and less antibiotic exposure, fecal inflammation, and diarrhea than Tox+/PCR+ patients (P < .001 for all). The median duration of diarrhea was shorter in Tox-/PCR+ patients (2 days; interquartile range, 1-4 days) than in Tox+/PCR+ patients (3 days; interquartile range, 1-6 days) (P = .003) and was similar to that in Tox-/PCR- patients (2 days; interquartile range, 1-3 days), despite minimal empirical treatment of Tox-/PCR+ patients. No CDI-related complications occurred in Tox-/PCR+ patients vs 10 complications in Tox+/PCR+ patients (0% vs 7.6%, P < .001). One Tox-/PCR+ patient had recurrent CDI as a contributing factor to death within 30 days vs 11 CDI-related deaths in Tox+/PCR+ patients (0.6% vs 8.4%, P = .001). CONCLUSIONS AND RELEVANCE Among hospitalized adults with suspected CDI, virtually all CDI-related complications and deaths occurred in patients with positive toxin immunoassay test results. Patients with a positive molecular test result and a negative toxin immunoassay test result had outcomes that were comparable to patients without C difficile by either method. Exclusive reliance on molecular tests for CDI diagnosis without tests for toxins or host response is likely to result in overdiagnosis, overtreatment, and increased health care costs.
Collapse
Affiliation(s)
- Christopher R Polage
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento2Division of Infectious Diseases, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento
| | - Clare E Gyorke
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento
| | - Michael A Kennedy
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento
| | - Jhansi L Leslie
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento3Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor
| | - David L Chin
- Center for Healthcare Policy and Research, University of California Davis, Sacramento
| | - Susan Wang
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento5Yolo County Health Department, Woodland, California
| | - Hien H Nguyen
- Division of Infectious Diseases, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento
| | - Bin Huang
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York7Department of Clinical Laboratory, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yi-Wei Tang
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York8Weill Medical College of Cornell University, New York, New York
| | - Lenora W Lee
- Division of Infectious Diseases, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento
| | - Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento
| | - Sandra Taylor
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento
| | - Patrick S Romano
- Center for Healthcare Policy and Research, University of California Davis, Sacramento10Division of General Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento11Division of General Pediatrics, Department
| | - Edward A Panacek
- Department of Emergency Medicine, University of California Davis School of Medicine, Sacramento
| | - Parker B Goodell
- Department of Emergency Medicine, University of California Davis School of Medicine, Sacramento
| | - Jay V Solnick
- Division of Infectious Diseases, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento13Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Sacramento
| | - Stuart H Cohen
- Division of Infectious Diseases, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento
| |
Collapse
|
47
|
Mao EJ, Kelly CR, Machan JT. Racial Differences in Clostridium difficile Infection Rates Are Attributable to Disparities in Health Care Access. Antimicrob Agents Chemother 2015; 59:6283-7. [PMID: 26248363 PMCID: PMC4576108 DOI: 10.1128/aac.00795-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/20/2015] [Indexed: 11/20/2022] Open
Abstract
This study confirms previously reported racial differences in Clostridium difficile infection (CDI) rates in the United States and explores the nature of those differences. We conducted a retrospective study using the 2010 Nationwide Inpatient Sample, the largest all-payer database of hospital discharges in the United States. We identified hospital stays most likely to include antibiotic treatment for infections, based on hospital discharge diagnoses, and we examined how CDI rates varied, in an attempt to distinguish between genotypic and environmental racial differences. Logistic regressions for the survey design were used to test hypotheses. Among patients likely to have received antibiotics, white patients had higher CDI rates than black, Hispanic, Asian, and Native American patients (P < 0.0001). CDI rates increased with higher income levels and were higher for hospitalizations paid by private insurance versus those paid by Medicaid or classified as self-pay or free care (P < 0.0001). Among patients admitted from skilled nursing facilities, where racial bias in health care access is less, racial differences in CDI rates disappeared (P = 1.0). Infected patients did not show racial differences in rates of complicated CDI or death (P = 1.0). Although white patients had greater CDI rates than nonwhite patients, racial differences in CDI rates disappeared in a population for which health care access was presumed to be less racially biased. This provides evidence that apparent racial differences in CDI risks may represent health care access disparities, rather than genotypic differences. CDI represents a deviation from the paradigm that increased health care access is associated with less morbidity.
Collapse
Affiliation(s)
- Eric J Mao
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA Division of Gastroenterology, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Colleen R Kelly
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA Division of Gastroenterology, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Jason T Machan
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA Lifespan Hospital System, Providence, Rhode Island, USA Departments of Orthopedics and Surgery, Rhode Island Hospital, Providence, Rhode Island, USA
| |
Collapse
|
48
|
Pilcante J, Rojas P, Ernst D, Sarmiento M, Ocqueteau M, Bertin P, García M, Rodriguez M, Jara V, Ajenjo M, Ramirez P. Clostridium difficile infection in Chilean patients submitted to hematopoietic stem cell transplantation. Rev Bras Hematol Hemoter 2015; 37:388-94. [PMID: 26670401 PMCID: PMC4678790 DOI: 10.1016/j.bjhh.2015.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/22/2015] [Accepted: 07/27/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction Patients submitted to hematopoietic stem cell transplantation have an increased risk of Clostridium difficile infection and multiple risk factors have been identified. Published reports have indicated an incidence from 9% to 30% of transplant patients however to date there is no information about infection in these patients in Chile. Methods A retrospective analysis was performed of patients who developed C. difficile infection after hematopoietic stem cell transplantations from 2000 to 2013. Statistical analysis used the Statistical Package for the Social Sciences software. Results Two hundred and fifty patients were studied (mean age: 39 years; range: 17–69), with 147 (59%) receiving allogeneic transplants and 103 (41%) receiving autologous transplants. One hundred and ninety-two (77%) patients had diarrhea, with 25 (10%) cases of C. difficile infection being confirmed. Twenty infected patients had undergone allogeneic transplants, of which ten had acute lymphoblastic leukemia, three had acute myeloid leukemia and seven had other diseases (myelodysplastic syndrome, chronic myeloid leukemia, severe aplastic anemia). In the autologous transplant group, five patients had C. difficile infection; two had multiple myeloma, one had amyloidosis, one had acute myeloid leukemia and one had germinal carcinoma. The overall incidence of C. difficile infection was 4% within the first week, 6.4% in the first month and 10% in one year, with no difference in overall survival between infected and non-infected groups (72.0% vs. 67.6%, respectively; p-value = 0.56). Patients infected after allogeneic transplants had a slower time to neutrophil engraftment compared to non-infected patients (17.5 vs. 14.9 days, respectively; p-value = 0.008). In the autologous transplant group there was no significant difference in the neutrophil engraftment time between infected and non-infected patients (12.5 days vs. 11.8 days, respectively; p-value = 0.71). In the allogeneic transplant group, the median time to acute graft-versus-host disease was similar between the two groups (p-value = 0.08), as was the incidence of grades 1–4 acute graft-versus-host disease (40% vs. 48%; p-value >0.05). Conclusion The incidence of C. difficile infection after hematopoietic stem cell transplantation was low, with a significant number of cases occurring shortly after transplantation. Allogeneic transplants had a three-time higher risk of infection compared to autologous transplants, but this was not associated with increased mortality, decreased overall survival or higher risk of acute graft-versus-host disease.
Collapse
Affiliation(s)
| | | | - Daniel Ernst
- Pontificia Universidad Católica, Santiago, Chile
| | | | | | - Pablo Bertin
- Pontificia Universidad Católica, Santiago, Chile
| | - Maria García
- Pontificia Universidad Católica, Santiago, Chile
| | | | | | - Maria Ajenjo
- Pontificia Universidad Católica, Santiago, Chile
| | | |
Collapse
|
49
|
Shields K, Araujo-Castillo RV, Theethira TG, Alonso CD, Kelly CP. Recurrent Clostridium difficile infection: From colonization to cure. Anaerobe 2015; 34:59-73. [PMID: 25930686 PMCID: PMC4492812 DOI: 10.1016/j.anaerobe.2015.04.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 12/16/2022]
Abstract
Clostridium difficile infection (CDI) is increasingly prevalent, dangerous and challenging to prevent and manage. Despite intense national and international attention the incidence of primary and of recurrent CDI (PCDI and RCDI, respectively) have risen rapidly throughout the past decade. Of major concern is the increase in cases of RCDI resulting in substantial morbidity, morality and economic burden. RCDI management remains challenging as there is no uniformly effective therapy, no firm consensus on optimal treatment, and reliable data regarding RCDI-specific treatment options is scant. Novel therapeutic strategies are critically needed to rapidly, accurately, and effectively identify and treat patients with, or at-risk for, RCDI. In this review we consider the factors implicated in the epidemiology, pathogenesis and clinical presentation of RCDI, evaluate current management options for RCDI and explore novel and emerging therapies.
Collapse
Affiliation(s)
- Kelsey Shields
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, United States.
| | - Roger V Araujo-Castillo
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Lowry Medical Office Building, Suite GB 110 Francis Street, Boston, MA 02215, United States.
| | - Thimmaiah G Theethira
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, United States.
| | - Carolyn D Alonso
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Lowry Medical Office Building, Suite GB 110 Francis Street, Boston, MA 02215, United States.
| | - Ciaran P Kelly
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, United States.
| |
Collapse
|
50
|
A Novel Quantitative Sampling Technique for Detection and Monitoring of Clostridium difficile Contamination in the Clinical Environment. J Clin Microbiol 2015; 53:2570-4. [PMID: 26041892 DOI: 10.1128/jcm.00376-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/26/2015] [Indexed: 01/08/2023] Open
Abstract
The horizontal transmission of Clostridium difficile in the hospital environment is difficult to establish. Current methods to detect C. difficile spores on surfaces are not quantitative, lack sensitivity, and are protracted. We propose a novel rapid method to detect and quantify C. difficile contamination on surfaces. Sponge swabbing was compared to contact plate sampling to assess the in vitro recovery of C. difficile ribotype 027 contamination (∼10(0), 10(1), or 10(2) CFU of spores) from test surfaces (a bed rail, a stainless steel sheet, or a polypropylene work surface). Sponge swab contents were concentrated by vacuum filtration, and the filter membrane was plated onto selective agar. The efficacy of each technique for the recovery of C. difficile from sites in the clinical environment that are touched at a high frequency was evaluated. Contact plates recovered 19 to 32% of the total contamination on test surfaces, whereas sponge swabs recovered 76 to 94% of the total contamination, and contact plates failed to detect C. difficile contamination below a detection limit of 10 CFU/25 cm(2) (0.4 CFU/cm(2)). In use, contact plates failed to detect C. difficile contamination (0/96 contact plates; 4 case wards), while sponge swabs recovered C. difficile from 29% (87/301) of the surfaces tested in the clinical environment. Approximately 74% (36/49) of the area in the vicinity of the patient was contaminated (∼1.34 ± 6.88 CFU/cm(2) C. difficile spores). Reservoirs of C. difficile extended to beyond the areas near the patient: a dirty utility room sink (2.26 ± 5.90 CFU/cm(2)), toilet floor (1.87 ± 2.40 CFU/cm(2)), and chair arm (1.33 ± 4.69 CFU/cm(2)). C. difficile was present on floors in ∼90% of case wards. This study highlights that sampling with a contact plate may fail to detect C. difficile contamination and result in false-negative reporting. Our sponge sampling technique permitted the rapid and quantitative measurement of C. difficile contamination on surfaces with a sensitivity (limit, 0 CFU) greater than that which is otherwise possible. This technique could be implemented for routine surface hygiene monitoring for targeted cleaning interventions and as a tool to investigate routes of patient-patient transmission in the clinical environment.
Collapse
|