1
|
Freeman J, Sanders IMJG, Harmanus C, Clark EV, Berry AM, Smits WK. Antimicrobial susceptibility testing of Clostridioides difficile: a dual-site study of three different media and three therapeutic antimicrobials. Clin Microbiol Infect 2025; 31:1011-1017. [PMID: 39884503 DOI: 10.1016/j.cmi.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
OBJECTIVES Increasing resistance to antimicrobials used for the treatment of Clostridioides difficile infections necessitates reproducible antimicrobial susceptibility testing. Current guidelines take a one-size-fits-all approach and/or offer limited guidance. We investigated how the choice of medium affects measured MIC values across two sites. METHODS We determined MIC values for the antimicrobials fidaxomicin, metronidazole, and vancomycin for a representative collection of European C. difficile strains (n = 235) using agar dilution on three different media: Brucella Blood Agar (BBA), Fastidious Anaerobe Agar supplemented with horse blood (FAA-HB), and Wilkins-Chalgren (WC) agar. The study was conducted at two sites to compare reproducibility. Usability (ease of preparation of the media as well as read-out of the assay) was assessed through a survey. RESULTS We found that all media result in highly consistent aggregated MIC data for all antibiotics, with MIC50 and MIC90 within two-fold of each other across sites. For fidaxomin, MIC values on WC were lower than on the other media (MIC90: WC = 0.125-0.25 mg/L; BBA and FAA-HB = 0.5 mg/L). Metronidazole showed the lowest MIC on BBA and the highest on WC (MIC90: WC = 2 mg/L; BBA = 0.5-1 mg/L; FAA-HB: 1-2 mg/L). For vancomycin, MIC values were similar across media (MIC90: all media = 1-2 mg/L). Though absolute values for individual isolates differed between sites, identified resistant isolates were similar. Results obtained on FAA-HB were most consistent between sites and results obtained on WC showed the most divergence. FAA-HB was positively evaluated in the usability survey. DISCUSSION This study shows medium-dependent differences in C. difficile MICs for at least two antimicrobials across two sites. We suggest the use of FAA-HB to align with general European Committee on Antimicrobial Susceptibility Testing (EUCAST) recommendations for susceptibility testing of anaerobic bacteria and deposited reference strains for standard susceptibility testing of C. difficile to increase interlaboratory reproducibility.
Collapse
Affiliation(s)
- Jane Freeman
- Healthcare Associated Infections Research Group, University of Leeds and Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom; European Society of Clinical Microbiology and Infectious Diseases Study Group on Clostridioides difficile, Basel, Switzerland.
| | - Ingrid M J G Sanders
- Experimental Bacteriology Research Group, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Céline Harmanus
- European Society of Clinical Microbiology and Infectious Diseases Study Group on Clostridioides difficile, Basel, Switzerland; Experimental Bacteriology Research Group, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Emma V Clark
- Healthcare Associated Infections Research Group, University of Leeds and Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Andrea M Berry
- Healthcare Associated Infections Research Group, University of Leeds and Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Wiep Klaas Smits
- European Society of Clinical Microbiology and Infectious Diseases Study Group on Clostridioides difficile, Basel, Switzerland; Experimental Bacteriology Research Group, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
2
|
Dirks EE, Pfiffer V, Sohl G, Berger FK, Friesen I, Friesen J, Ignatius R, Elias J, Mellmann A, Arvand M. Whole genome sequence analysis reveals limited diversity among Clostridioides difficile ribotype 027 and 078 isolates collected in 22 hospitals in Berlin and Brandenburg, Germany. Antimicrob Resist Infect Control 2025; 14:56. [PMID: 40437588 PMCID: PMC12121276 DOI: 10.1186/s13756-025-01565-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 05/04/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Clostridioides difficile infections (CDI) present significant health risks and are among the most important nosocomial infections. Ribotype (RT) 027 poses a particular risk due to its proposed "hypervirulence". Traditionally, C. difficile isolates are characterized using PCR-ribotyping. More recently, whole genome sequence (WGS) analysis is increasingly used, which may provide a higher discriminatory power. This study aimed to assess the distribution of different C. difficile RTs in hospitals in the Berlin-Brandenburg area, and to analyse the heterogeneity within isolates of different ribotypes using WGS. METHODS Between February 2020 and November 2021, stool samples from patients with laboratory-confirmed CDI were collected from 22 hospitals (approximately 13,900 beds) in Berlin and Brandenburg. Toxigenic isolates (n = 476) were further characterized by ribotyping, antibiotic susceptibility testing, toxinotyping, and core genome multilocus sequence typing (cgMLST). RESULTS Sixty-five different RTs were detected, with RT014 (16.1%), RT027 (12.8%), and RT001 (7.6%) being the most prevalent. RT027 isolates exhibited resistance to several antibiotics. Further, cgMLST analysis revealed very close genetic relatedness between RT027 isolates despite being epidemiologically unrelated. Similar findings of a monomorphic population were observed for RT078 isolates. In contrast, other RTs showed a heterogenic population structure. CONCLUSIONS This study provides first insights into the distribution of C. difficile genotypes, corresponding antimicrobial resistance, and clonal relatedness using cgMLST, highlighting RT027 as the second most common genotype for the studied area. For the monomorphic RT027 and RT078 populations, new definitions of clonal relatedness might be necessary.
Collapse
Affiliation(s)
- Esther E Dirks
- Unit Hospital Hygiene, Infection Prevention and Control, Department for Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Vanessa Pfiffer
- Unit Hospital Hygiene, Infection Prevention and Control, Department for Infectious Diseases, Robert Koch Institute, Berlin, Germany
- Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Genevieve Sohl
- Unit Hospital Hygiene, Infection Prevention and Control, Department for Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Fabian K Berger
- National Reference Center for Clostridioides Difficile, Homburg Branch, Homburg/Saar, Germany
- Institute of Virology, Saarland University Medical Center, Homburg/Saar, Germany
| | - Ina Friesen
- Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
- National and WHO Supranational Reference Center for Mycobacteria, Research Center Borstel, Borstel, Germany
| | | | - Ralf Ignatius
- Medizinisches Versorgungszentrum Labor 28 GmbH, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Berlin, Berlin, Germany
| | - Johannes Elias
- DRK Kliniken Berlin Westend, Berlin, Germany
- HMU Health and Medical University GmbH, Potsdam, Germany
| | - Alexander Mellmann
- Institute of Hygiene, University Hospital Muenster and National Reference Center for Clostridioides Difficile, Münster Branch, Münster, Germany
| | - Mardjan Arvand
- Unit Hospital Hygiene, Infection Prevention and Control, Department for Infectious Diseases, Robert Koch Institute, Berlin, Germany.
- Institute for Medical Microbiology and Hygiene, Department of Infectious Diseases, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
3
|
Sauvat L, Verhoeven PO, Gagnaire J, Berthelot P, Paul S, Botelho-Nevers E, Gagneux-Brunon A. Vaccines and monoclonal antibodies to prevent healthcare-associated bacterial infections. Clin Microbiol Rev 2024; 37:e0016022. [PMID: 39120140 PMCID: PMC11391692 DOI: 10.1128/cmr.00160-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
SUMMARYHealthcare-associated infections (HAIs) represent a burden for public health with a high prevalence and high death rates associated with them. Pathogens with a high potential for antimicrobial resistance, such as ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) and Clostridioides difficile, are responsible for most HAIs. Despite the implementation of infection prevention and control intervention, globally, HAIs prevalence is stable and they are mainly due to endogenous pathogens. It is undeniable that complementary to infection prevention and control measures, prophylactic approaches by active or passive immunization are needed. Specific groups at-risk (elderly people, chronic condition as immunocompromised) and also healthcare workers are key targets. Medical procedures and specific interventions are known to be at risk of HAIs, in addition to hospital environmental exposure. Vaccines or monoclonal antibodies can be seen as attractive preventive approaches for HAIs. In this review, we present an overview of the vaccines and monoclonal antibodies in clinical development for prevention of the major bacterial HAIs pathogens. Based on the current state of knowledge, we look at the challenges and future perspectives to improve prevention by these means.
Collapse
Affiliation(s)
- Léo Sauvat
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Paul O Verhoeven
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Agents and Hygiene, University-Hospital of Saint-Etienne, Saint-Etienne, France
| | - Julie Gagnaire
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Philippe Berthelot
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Infection Control Unit, University Hospital of Saint-Etienne, Saint-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Elisabeth Botelho-Nevers
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Amandine Gagneux-Brunon
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, Université Jean Monnet St-Etienne, St-Etienne, France
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC 1408 Inserm, Axe vaccinologie, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
4
|
Antunes A, Tricotel A, Wilk A, Dombrowski S, Rinta-Kokko H, Andersson FL, Ghosh S. Estimating excess mortality and economic burden of Clostridioides difficile infections and recurrences during 2015-2019: the RECUR Germany study. BMC Infect Dis 2024; 24:548. [PMID: 38822244 PMCID: PMC11143700 DOI: 10.1186/s12879-024-09422-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Clostridioides difficile infections (CDIs) and recurrences (rCDIs) remain a major public health challenge due to substantial mortality and associated costs. This study aims to generate real-world evidence on the mortality and economic burden of CDI in Germany using claims data between 2015 and 2019. METHODS A longitudinal and matched cohort study using retrospective data from Statutory Health Insurance (SHI) was conducted in Germany with the BKK database. Adults diagnosed with CDI in hospital and community settings between 2015 and 2018 were included in the study. Patients had a minimum follow-up of 12-months. All-cause mortality was described at 6-, 12-, and 24-months. Healthcare resource usage (HCRU) and associated costs were assessed at 12-months of follow-up. A cohort of non-CDI patients matched by demographic and clinical characteristics was used to assess excess mortality and incremental costs of HCRU. Up to three non-CDI patients were matched to each CDI patient. RESULTS A total of 9,977 CDI patients were included in the longitudinal cohort. All-cause mortality was 32%, 39% and 48% at 6-, 12-, and 24-months, respectively, with minor variations by number of rCDIs. When comparing matched CDI (n = 5,618) and non-CDI patients (n = 16,845), CDI patients had an excess mortality of 2.17, 1.35, and 0.94 deaths per 100 patient-months, respectively. HCRU and associated costs were consistently higher in CDI patients compared to non-CDI patients and increased with recurrences. Total mean and median HCRU cost per patient during follow-up was €12,893.56 and €6,050 in CDI patients, respectively, with hospitalisations representing the highest proportion of costs. A total mean incremental cost per patient of €4,101 was estimated in CDI patients compared to non-CDI patients, increasing to €13,291 in patients with ≥ 3 rCDIs. CONCLUSIONS In this real-world study conducted in Germany, CDI was associated with increased risk of death and substantial costs to health systems due to higher HCRU, especially hospitalisations. HCRU and associated costs were exacerbated by rCDIs.
Collapse
Affiliation(s)
- Ana Antunes
- IQVIA, Global Database Studies, Real World Solutions, Edifício 3, Lagoas Park, Oeiras, Lisboa, 2740 - 266, Portugal.
| | | | - Adrian Wilk
- Team Gesundheit, Gesellschaft für Gesundheitsmanagement mbH, Essen, Germany
| | | | - Hanna Rinta-Kokko
- IQVIA, Global Database Studies, Real World Solutions, Espoo, Finland
| | | | - Subrata Ghosh
- College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
5
|
Ghosh S, Antunes A, Rinta-Kokko H, Chaparova E, Lay-Flurrie S, Tricotel A, Andersson FL. Estimating excess mortality and economic burden of Clostridioides difficile infections and recurrences during 2015-2019: The RECUR England study. Int J Infect Dis 2024; 142:106967. [PMID: 38368927 DOI: 10.1016/j.ijid.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024] Open
Abstract
OBJECTIVES To generate real-world evidence on all-cause mortality and economic burden of Clostridioides difficile infections (CDIs) and recurrences (rCDIs) in England. METHODS We conducted a cohort study using retrospective data from Clinical Practice Research Datalink linked to Hospital Episode Statistics. Patients diagnosed with CDI in hospital and community settings during 2015-2018 were included and followed for ≥1 year. All-cause mortality was described at 6, 12, and 24 months. Healthcare resource usage (HCRU) and associated costs were assessed at 12 months of follow-up. A cohort of non-CDI patients, matched by demographic and clinical characteristics including Charlson Comorbidity Index score, was used to assess excess mortality and incremental costs of HCRU. RESULTS All-cause mortality among CDI patients at 6, 12, and 24 months was 15.87%, 20.37%, and 27.03%, respectively. A higher proportion of rCDI patients died at any point during follow-up. Compared with matched non-CDI patients, excess mortality was highest at 6 months with 1.81 and 2.53 deaths per 100 patient-months among CDI and ≥1 rCDI patients. Hospitalizations were the main drivers of costs, with an incremental cost of £1209.21 per CDI patient. HCRU and costs increased with rCDIs. CONCLUSION CDI poses a substantial mortality and economic burden, further amplified by rCDIs.
Collapse
Affiliation(s)
- Subrata Ghosh
- College of Medicine and Health, University College Cork, Cork, Ireland; University of Birmingham, Birmingham, United Kingdom
| | - Ana Antunes
- IQVIA, Global Database Studies, Real World Solutions, Lisbon, Portugal.
| | - Hanna Rinta-Kokko
- IQVIA, Global Database Studies, Real World Solutions, Espoo, Finland
| | | | | | | | | |
Collapse
|
6
|
Buddle JE, Fagan RP. Pathogenicity and virulence of Clostridioides difficile. Virulence 2023; 14:2150452. [PMID: 36419222 DOI: 10.1080/21505594.2022.2150452] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Clostridioides difficile is the most common cause of nosocomial antibiotic-associated diarrhea, and is responsible for a spectrum of diseases characterized by high levels of recurrence, morbidity, and mortality. Treatment is complex, since antibiotics constitute both the main treatment and the major risk factor for infection. Worryingly, resistance to multiple antibiotics is becoming increasingly widespread, leading to the classification of this pathogen as an urgent threat to global health. As a consummate opportunist, C. difficile is well equipped for promoting disease, owing to its arsenal of virulence factors: transmission of this anaerobe is highly efficient due to the formation of robust endospores, and an array of adhesins promote gut colonization. C. difficile produces multiple toxins acting upon gut epithelia, resulting in manifestations typical of diarrheal disease, and severe inflammation in a subset of patients. This review focuses on such virulence factors, as well as the importance of antimicrobial resistance and genome plasticity in enabling pathogenesis and persistence of this important pathogen.
Collapse
Affiliation(s)
- Jessica E Buddle
- Molecular Microbiology, School of Biosciences, University of Sheffield, Sheffield, UK
| | - Robert P Fagan
- Molecular Microbiology, School of Biosciences, University of Sheffield, Sheffield, UK
| |
Collapse
|
7
|
Tossens B, Barthelme P, Briquet C, Belkhir L, Ngyuvula E, Soumillion K, Verroken A, Rodriguez-Villalobos H, Delmée M, Anantharajah A. Impact of the COVID-19 pandemic on Clostridioides difficile infection in a tertiary healthcare institution in Belgium. Acta Clin Belg 2023; 78:459-466. [PMID: 37608759 DOI: 10.1080/17843286.2023.2250624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/17/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVES Clostridioides difficile infection (CDI) causes the greatest number of healthcare-associated infectious diarrhoea. CDIs are transmitted by direct and indirect patient-to-patient contact and risk increases with the use of antibiotics. Since early 2020, the COVID-19 pandemic has affected healthcare systems in many ways including substantial changes in hygiene behaviour. The aim of this study was to assess whether CDI incidence differed during the COVID-19 pandemic compared to a year before. METHODS All tests for suspected CDI cases were recorded for a hospital in Brussels, Belgium. The percentage of CDI-positive results and incidences (total and healthcare-associated (HA)-CDI)) for years 2019, 2020, 2021, and 2022 were calculated. Antibiotic consumption was analysed for years 2019 and 2020. RESULTS Since the COVID-19 pandemic struck, a significant reduction of up to 39% was observed in the number of Clostridioides difficile stool tests in our hospital. A significant decrease in the percentage of positive tests and a 50% decrease in the incidence of CDI (total and HA-CDI) was found for 2020 compared with 2019 and confirmed for years 2021 and 2022. The decrease in CDI incidence was mostly marked in haematology, nephrology, and gastroenterology units. No significant change in the use of antibiotics was found. CONCLUSION The global decrease in CDI incidence observed in our hospital was not associated with a change in the use of antibiotics. The control measures implemented to prevent COVID-19 transmission may explain a reduction in CDI incidence. An underdiagnosis of CDI cannot be excluded.
Collapse
Affiliation(s)
- Bastien Tossens
- Department of Clinical Microbiology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Philippe Barthelme
- Department of Pharmacy, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Caroline Briquet
- Department of Pharmacy, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Leila Belkhir
- Department of Internal Medicine and Infectious Diseases, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Eléonore Ngyuvula
- National Reference Center Clostridioides difficile, Microbiology Unit, Université Catholique de Louvain, Brussels, Belgium
| | - Kate Soumillion
- National Reference Center Clostridioides difficile, Microbiology Unit, Université Catholique de Louvain, Brussels, Belgium
| | - Alexia Verroken
- Department of Clinical Microbiology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | - Michel Delmée
- National Reference Center Clostridioides difficile, Microbiology Unit, Université Catholique de Louvain, Brussels, Belgium
| | - Ahalieyah Anantharajah
- Department of Clinical Microbiology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- National Reference Center Clostridioides difficile, Microbiology Unit, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
8
|
Brauns S, Marquardt I, Thon C, Frentzel S, Jakob J, Färber J, Philipsen L, Jänsch L, Link A, Bruder D. Mucosal-associated invariant T cells from Clostridioides difficile-infected patients exhibit a distinct proinflammatory phenotype and enhanced cytotoxic activity. Int Immunol 2023; 35:543-554. [PMID: 37549964 DOI: 10.1093/intimm/dxad032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/07/2023] [Indexed: 08/09/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells mainly found in the mucosa and peripheral blood. We have recently demonstrated that Clostridioides difficile activates MAIT cells in vitro. However, their role in the pathogenesis of C. difficile infection (CDI) in human patients remains elusive to date. In this study, we performed comprehensive immunophenotyping of MAIT cells derived from CDI patients and compared their phenotype to that of patients with inflammatory bowel diseases (IBD) and healthy controls. Our study revealed that blood MAIT cells from CDI patients exhibit an interleukin 17a (IL-17a)-dominated proinflammatory phenotype and an increased readiness to synthesize the proinflammatory cytokine interferon γ (IFN-γ) following in vitro re-stimulation. Moreover, the cytotoxic activity of MAIT cells, as measured by surface CD107a and intracellular granzyme B expression, was strongly increased in CDI. Multi epitope ligand cartography (MELC) analysis of intestinal biopsies from CDI patients revealed that MAIT cells exhibit an increased production of granzyme B and increased cytotoxicity compared to the control group. Together with previously published in vitro data from our group, our findings suggest that MAIT cells are functionally involved in the immune response against C. difficile and contribute to the pathogenesis of CDI.
Collapse
Affiliation(s)
- Steffen Brauns
- Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Isabel Marquardt
- Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Cosima Thon
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Sarah Frentzel
- Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Josefine Jakob
- Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jacqueline Färber
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Lars Philipsen
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Multi-parametric Bioimaging and Cytometry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Lothar Jänsch
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | - Dunja Bruder
- Infection Immunology, Institute of Medical Microbiology and Hospital Hygiene, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
9
|
Vázquez-Cuesta S, Lozano García N, Fernández AI, Olmedo M, Kestler M, Alcalá L, Marín M, Bermejo J, Díaz FFA, Muñoz P, Bouza E, Reigadas E. Microbiome profile and calprotectin levels as markers of risk of recurrent Clostridioides difficile infection. Front Cell Infect Microbiol 2023; 13:1237500. [PMID: 37780848 PMCID: PMC10534046 DOI: 10.3389/fcimb.2023.1237500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Clostridioides difficile infection (CDI) is the main cause of nosocomial diarrhoea in developed countries. Recurrent CDI (R-CDI), which affects 20%-30% of patients and significantly increases hospital stay and associated costs, is a key challenge. The main objective of this study was to explore the role of the microbiome and calprotectin levels as predictive biomarkers of R-CDI. Methods We prospectively (2019-2021) included patients with a primary episode of CDI. Clinical data and faecal samples were collected. The microbiome was analysed by sequencing the hypervariable V4 region of the 16S rRNA gene on an Illumina Miseq platform. Results We enrolled 200 patients with primary CDI, of whom 54 developed R-CDI and 146 did not. We analysed 200 primary samples and found that Fusobacterium increased in abundance, while Collinsella, Senegalimassilia, Prevotella and Ruminococcus decreased in patients with recurrent versus non-recurrent disease. Elevated calprotectin levels correlated significantly with R-CDI (p=0.01). We built a risk index for R-CDI, including as prognostic factors age, sex, immunosuppression, toxin B amplification cycle, creatinine levels and faecal calprotectin levels (overall accuracy of 79%). Discussion Calprotectin levels and abundance of microbial genera such as Fusobacterium and Prevotella in primary episodes could be useful as early markers of R-CDI. We propose a readily available model for prediction of R-CDI that can be applied at the initial CDI episode. The use of this tool could help to better tailor treatments according to the risk of R-CDI.
Collapse
Affiliation(s)
- Silvia Vázquez-Cuesta
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Biochemistry and Molecular Biology Department, Faculty of Biology, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Nuria Lozano García
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ana I. Fernández
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - María Olmedo
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Martha Kestler
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Luis Alcalá
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Mercedes Marín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Javier Bermejo
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Fernández-Avilés Díaz
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Elena Reigadas
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| |
Collapse
|
10
|
Herzog MKM, Cazzaniga M, Peters A, Shayya N, Beldi L, Hapfelmeier S, Heimesaat MM, Bereswill S, Frankel G, Gahan CG, Hardt WD. Mouse models for bacterial enteropathogen infections: insights into the role of colonization resistance. Gut Microbes 2023; 15:2172667. [PMID: 36794831 PMCID: PMC9980611 DOI: 10.1080/19490976.2023.2172667] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
Globally, enteropathogenic bacteria are a major cause of morbidity and mortality.1-3 Campylobacter, Salmonella, Shiga-toxin-producing Escherichia coli, and Listeria are among the top five most commonly reported zoonotic pathogens in the European Union.4 However, not all individuals naturally exposed to enteropathogens go on to develop disease. This protection is attributable to colonization resistance (CR) conferred by the gut microbiota, as well as an array of physical, chemical, and immunological barriers that limit infection. Despite their importance for human health, a detailed understanding of gastrointestinal barriers to infection is lacking, and further research is required to investigate the mechanisms that underpin inter-individual differences in resistance to gastrointestinal infection. Here, we discuss the current mouse models available to study infections by non-typhoidal Salmonella strains, Citrobacter rodentium (as a model for enteropathogenic and enterohemorrhagic E. coli), Listeria monocytogenes, and Campylobacter jejuni. Clostridioides difficile is included as another important cause of enteric disease in which resistance is dependent upon CR. We outline which parameters of human infection are recapitulated in these mouse models, including the impact of CR, disease pathology, disease progression, and mucosal immune response. This will showcase common virulence strategies, highlight mechanistic differences, and help researchers from microbiology, infectiology, microbiome research, and mucosal immunology to select the optimal mouse model.
Collapse
Affiliation(s)
- Mathias K.-M. Herzog
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Monica Cazzaniga
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Audrey Peters
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Nizar Shayya
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Luca Beldi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | - Markus M. Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Gad Frankel
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Cormac G.M. Gahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Vázquez-Cuesta S, Villar L, García NL, Fernández AI, Olmedo M, Alcalá L, Marín M, Muñoz P, Bouza E, Reigadas E. Characterization of the gut microbiome of patients with Clostridioides difficile infection, patients with non- C. difficile diarrhea, and C. difficile-colonized patients. Front Cell Infect Microbiol 2023; 13:1130701. [PMID: 37124040 PMCID: PMC10130453 DOI: 10.3389/fcimb.2023.1130701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Clostridioides difficile infection (CDI) is the main cause of nosocomial diarrhea in developed countries. A key challenge in CDI is the lack of objective methods to ensure more accurate diagnosis, especially when differentiating between true infection and colonization/diarrhea of other causes. The main objective of this study was to explore the role of the microbiome as a predictive biomarker of CDI. Methods Between 2018 and 2021, we prospectively included patients with CDI, recurrent CDI (R-CDI), non-CDI diarrhea (NO-CDI), colonization by C. difficile, and healthy individuals. Clinical data and fecal samples were collected. The microbiome was analyzed by sequencing the hypervariable V4 region of the 16S rRNA gene on an Illumina Miseq platform. The mothur bioinformatic pipeline was followed for pre-processing of raw data, and mothur and R were used for data analysis. Results During the study period, 753 samples from 657 patients were analyzed. Of these, 247 were from patients with CDI, 43 were from patients colonized with C. difficile, 63 were from healthy individuals, 324 were from NOCDI, and 76 were from R-CDI. We found significant differences across the groups in alpha and beta diversity and in taxonomic abundance. We identified various genera as the most significant biomarkers for CDI (Bacteroides, Proteus, Paraprevotella, Robinsoniella), R-CDI (Veillonella, Fusobacterium, Lactobacillus, Clostridium sensu stricto I), and colonization by C. difficile (Parabacteroides, Faecalicoccus, Flavonifractor, Clostridium XVIII). Discussion We observed differences in microbiome patterns between healthy individuals, colonized patients, CDI, R-CDI, and NOCDI diarrhea. We identified possible microbiome biomarkers that could prove useful in the diagnosis of true CDI infections. Further studies are warranted.
Collapse
Affiliation(s)
- Silvia Vázquez-Cuesta
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Biochemistry and Molecular Biology Department, Faculty of Biology, Universidad Complutense de Madrid (UCM), Madrid, Spain
- *Correspondence: Silvia Vázquez-Cuesta,
| | - Laura Villar
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Nuria Lozano García
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ana I. Fernández
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - María Olmedo
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Luis Alcalá
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Mercedes Marín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Clostridioides difficile (ESGCD), Basel, Switzerland
| | - Elena Reigadas
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Clostridioides difficile (ESGCD), Basel, Switzerland
| |
Collapse
|
12
|
Influence of Binary Toxin Gene Detection and Decreased Susceptibility to Antibiotics among Clostridioides difficile Strains on Disease Severity: a Single-Center Study. Antimicrob Agents Chemother 2022; 66:e0048922. [PMID: 35861541 PMCID: PMC9380565 DOI: 10.1128/aac.00489-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Clostridioides difficile infection (CDI) is the fifth leading cause of death from nonmalignant gastrointestinal disease in the United States. The contribution of resistance to C. difficile-active antibiotics to the outcomes of CDI is unclear. We evaluated the antimicrobial susceptibility of C. difficile isolates in a U.S. hospital and determined associations of clinical variables and binary toxin positivity with antibiotic resistance. C. difficile spores were cultured from fecal specimens of adult patients with CDI for genotyping and antimicrobial susceptibility assay (for clindamycin [CLI], fidaxomicin [FDX], metronidazole [MTZ], moxifloxacin [MXF], tigecycline [TGC], and vancomycin [VAN]). Electronic medical records were reviewed for clinical data extraction. Ninety-seven of 130 (75%) fecal samples grew toxigenic C. difficile in culture. Most of the isolates were tcdA+ tcdB+ cdtB- (80.4%), and 18.6% and 1% were tcdA+ tcdB+ cdtB+ and tcdA-tcdB+ cdtB+, respectively. Susceptibility to VAN, MTZ, FDX, TGC, MXF, and CLI was 96%, 94%, 100%, 100%, 8%, and 79%, respectively. Six isolates, all cdtB positive and belonging to the 027 ribotype, were resistant to VAN and/or MTZ. Higher MICs were found in isolates with a mutation in the VAN-related resistance gene vanR, but not vanS. In addition, cdtB+ isolates exhibited higher MICs of VAN, MTZ, TGC, CLI, and MXF compared to cdtB- strains. Patients with greater intestinal inflammation or severe disease were more likely to be infected with cdtB+ strains. Decreased susceptibility to antibiotics is not directly associated with either severe or recurrent CDI. However, antimicrobial susceptibility of C. difficile is decreased in strains positive for the binary toxin gene.
Collapse
|
13
|
Olmedo M, Valerio M, Reigadas E, Marín M, Alcalá L, Muñoz P, Bouza E. Clinical impact of a Clostridioides ( Clostridium) difficile bedside infectious disease stewardship intervention. JAC Antimicrob Resist 2020; 2:dlaa037. [PMID: 34223003 PMCID: PMC8210181 DOI: 10.1093/jacamr/dlaa037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/23/2020] [Accepted: 04/06/2020] [Indexed: 11/17/2022] Open
Abstract
Objectives To evaluate the clinical impact of a bedside visit to patients with a positive Clostridioides difficile test on the antimicrobial stewardship of C. difficile infection (CDI) and non-C. difficile infections. Methods All patients ≥18 years old with positive CDI laboratory tests hospitalized between January 2017 and August 2017 received an immediate bedside intervention that consisted mainly of checking protective measures and providing recommendations on infection control and the management of CDI and other infections. Results A total of 214 patients were evaluated. The infectious disease (ID) physician was the first to establish protective measures in 25.2% of the cases. In 22/29 (75.9%) cases, physicians in charge accepted ID consultant recommendations to stop CDI treatment in asymptomatic patients. Unnecessary non-CDI antibiotics were discontinued in 19.1% of the cases. ID recommendations were not accepted by physicians in charge in only 12.6% of the cases. Conclusions A bedside rapid intervention for patients with a CDI-positive faecal sample was effective in avoiding overdiagnosis and unnecessary antibiotic treatment, optimizing anti-CDI drugs, increasing compliance with infection control measures and providing educational advice.
Collapse
Affiliation(s)
- María Olmedo
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Maricela Valerio
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Elena Reigadas
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Mercedes Marín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Luis Alcalá
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
14
|
Robertson C, Pan J, Kavanagh K, Ford I, McCowan C, Bennie M, Marwick C, Leanord A. Cost burden of Clostridioides difficile infection to the health service: A retrospective cohort study in Scotland. J Hosp Infect 2020; 106:554-561. [PMID: 32717202 DOI: 10.1016/j.jhin.2020.07.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/15/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Clostridioides difficile infection (CDI) is associated with high healthcare demands and related costs. AIM To evaluate the healthcare and economic burden of CDI in hospitalized patients with community- (HOCA-CDI) or hospital-associated CDI (HOHA-CDI) in the National Health Service in Scotland. METHODS A retrospective cohort study was conducted, examining data between August 2010 and July 2013 from four patient-level Scottish datasets, linked to death data. Data examined included prior antimicrobial prescriptions in the community, hospitalizations, length of stay and mortality. Each CDI case was matched to three hospital-based controls on the basis of age, gender, hospital and date of admission. Descriptive economic evaluations were based on bed-day costs for different types of wards. FINDINGS Overall, 3304 CDI cases were included in the study. CDI was associated with additional median lengths of stay of 7.2 days for HOCA-CDI and 12.0 days for HOHA-CDI compared with their respective, matched controls. The 30-day mortality rate was 6.8% for HOCA-CDI and 12.4% for HOHA-CDI. Overall, recurrence within 90 days of the first CDI episode occurred in 373/2740 (13.6%) survivors. The median additional expenditure for each initial CDI case compared with matched controls was £1713. In the 6 months after the index hospitalization, the cost associated with a CDI case was £5126 higher than for controls. CONCLUSION Using routinely collected national data, we demonstrated the substantial burden of CDI on healthcare services, including lengthy hospital stays and readmissions, which increased the costs of managing patients with CDI compared with matched controls.
Collapse
Affiliation(s)
- C Robertson
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK; Health Protection Scotland, NHS National Services Scotland, Glasgow, UK
| | - J Pan
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
| | - K Kavanagh
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
| | - I Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - C McCowan
- School of Medicine, University of St Andrews, St Andrews, UK
| | - M Bennie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK; Public Health and Intelligence, NHS National Services Scotland, Edinburgh, UK
| | - C Marwick
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - A Leanord
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK; NHS Greater Glasgow and Clyde, Glasgow, UK.
| |
Collapse
|
15
|
Jhaveri K, Som A, Padala SA, Surani S. The Impact of Clostridium Difficile Infections on In-Hospital Outcomes of Venous Thromboembolism (Deep Vein Thrombosis or Pulmonary Embolism) Hospitalizations. Cureus 2020; 12:e9195. [PMID: 32685328 PMCID: PMC7366047 DOI: 10.7759/cureus.9195] [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: 12/03/2022] Open
Abstract
Background Clostridium difficile infection (CDI) is associated with high mortality. Studies have shown an increased rate of venous thromboembolism (VTE) in patients with CDI. However, literature regarding the impact of CDI on outcomes of VTE-related hospitalizations is scarce. Our study aimed to assess the impact of CDI on in-hospital outcomes among VTE hospitalizations. Methods The 2016 National Inpatient Sample (NIS) was used to identify all adult hospitalizations in the United States with a primary discharge diagnosis of acute VTE. Hospitalizations with deep vein thrombosis (DVT) or pulmonary embolism (PE) were included under VTE. The sample was stratified based on the presence or absence of active CDI. Chi-square test and weighted Student’s t-test were used to analyze categorical and continuous variables, respectively. The adjusted odds ratio (OR) for clinical outcomes were calculated using multivariate logistic regression analysis. Subgroup analyses for DVT and PE hospitalizations were performed. All analyses were completed in SAS (SAS Institute Inc., Cary, NC), and a p-value of <0.05 was considered statistically significant. Results We identified 382,585 weighted hospitalizations for VTE. Among them, 0.8% had concomitant CDI. The presence of CDI was associated with a statistically significant increase in in-hospital mortality (6% vs. 3%), hospitalization cost ($147,356.5 vs. $55,193), and length of stay (13.7 vs. 5.4 days). There were more incidents of bleeding and acute respiratory failure requiring prolonged ventilation in patients with CDI. The odds of stroke were significantly higher in patients with CDI and DVT. Conclusion CDI independently increased in-hospital mortality in VTE. Preventing CDI in the VTE population may mitigate complications, improve in-hospital outcomes, and reduce treatment costs.
Collapse
|
16
|
Okumura H, Ueyama M, Shoji S, English M. Cost-effectiveness analysis of fidaxomicin for the treatment of Clostridioides (Clostridium) difficile infection in Japan. J Infect Chemother 2020; 26:611-618. [PMID: 32165072 DOI: 10.1016/j.jiac.2020.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/15/2020] [Accepted: 01/31/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The cost of treating Clostridioides difficile infection (CDI), particularly recurrent disease, is high. In clinical trials, fidaxomicin has been associated with significantly lower recurrence rates and higher sustained cure rates versus vancomycin. The high acquisition cost of fidaxomicin has limited its acceptance into clinical practice. OBJECTIVE To evaluate the cost-effectiveness of fidaxomicin versus vancomycin in patients with CDI after failure of metronidazole in the Japanese healthcare setting. METHODS Clinical results from three phase III trials and inputs based on assumptions validated by clinical experts in Japan were used in a semi-Markov model with 1-year time horizon. Incremental cost-effectiveness ratios (ICERs) for fidaxomicin versus vancomycin were expressed as cost per quality-adjusted life year (QALY) and interpreted using willingness-to-pay thresholds of JPY 5,000,000 (primary) and JPY 7,500,000 (secondary) per QALY gained in Japan. Probabilistic sensitivity analyses and scenario analyses were performed. RESULTS Higher drug acquisition costs for fidaxomicin were partially offset by lower hospitalization costs driven by fewer recurrences, lower costs of complications, and fewer general practitioner visits versus vancomycin. The ICER for fidaxomicin versus vancomycin was estimated at JPY 5,715,183 per QALY gained. Sensitivity analyses showed a 46% probability of fidaxomicin being cost-effective versus vancomycin at a willingness-to-pay threshold of JPY 5,000,000 per QALY gained. At a threshold of JPY 7,500,000, there was a 54% probability of fidaxomicin being cost-effective. CONCLUSIONS Fidaxomicin treatment in patients with CDI following failure of metronidazole improves health outcomes with partial offset of higher drug acquisition costs versus vancomycin.
Collapse
Affiliation(s)
| | | | - Shingo Shoji
- Medical Affairs, Astellas Pharma, Inc., Tokyo, Japan.
| | - Marci English
- Astellas Pharma Global Development, Inc., Northbrook, IL, USA.
| |
Collapse
|
17
|
A systematic review of economic evaluation in fecal microbiota transplantation. Infect Control Hosp Epidemiol 2020; 41:458-466. [DOI: 10.1017/ice.2019.371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AbstractBackground:Fecal microbiota transplantation (FMT) is an effective therapy in recurrent Clostridium difficile infection (rCDI). It is only recommended for this indication by European and American guidelines. Other indications of FMT are being studied, such as inflammatory bowel disease (IBD), and they have shown promising results.Objectives:To identify and review published FMT-related economic evaluations (EEs) to assess their quality and the economic impact of FMT in the treatment of these diseases.Data sources:The systematic literature research was conducted in both PubMed and Cochrane to identify EEs published before July 1, 2019.Study eligibility criteria:Articles were included if they concerned FMT (whatever the disease and its line of treatment), if they reported full or partial EEs, and if they were written in English. Articles were excluded if they did not concern FMT; if they did not report an EE; or if they were a systematic review, editorial, comment, letter to the editor, practice point, or poster.Methods:A measurement tool, AMSTAR, was used to optimize the quality of this systematic review. Based on the CHEERS checklist, data were identified and extracted from articles. The quality of each EE was assessed using the Drummond checklist.Results:Overall, 9 EEs were included: all EEs were full evaluations and 8 were cost-utility analyses (CUAs). All EEs had a Drummond score ≥ 7, which indicated high quality. All CUAs related to rCDI and IBD concluded that FMT was cost-effective compared with other reference treatments, at a threshold ≤$50,000/QALY. One EE about initial CDI showed that FMT was dominated by metronidazole.Conclusions:Despite a limited number of EEs, FMT seems to be a promising and cost-effective treatment for rCDI. More EE studies on other diseases like IBD are necessary to address FMT efficiency for new indications. Therefore, our systematic review provides a framework for healthcare decision making.
Collapse
|
18
|
Kelly A, Salgado PS. The engulfasome in C. difficile: Variations on protein machineries. Anaerobe 2019; 60:102091. [PMID: 31470088 PMCID: PMC6934232 DOI: 10.1016/j.anaerobe.2019.102091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 08/18/2019] [Accepted: 08/22/2019] [Indexed: 12/26/2022]
Abstract
Clostridioides difficile infection (CDI) continues to be a substantial healthcare burden, and the changing disease profile raises new challenges in CDI management, both in clinical settings and in the community. CDI is transmitted by spores, which are formed by a subset of the cell population where an asymmetric septum is formed. A full copy of the chromosome is transported into the smaller compartment which is then engulfed by the mother cell. After engulfment, multiple metabolic and morphological changes occur, eventually resulting in the release of the mature spore. Whilst studies in the model organism Bacillus subtilis have demonstrated the importance of the DMP and Q:AH machineries in engulfment, it is becoming clear that there are fundamental differences in the way the two organisms organise these machineries. As spores are the infectious agent in CDI, it is crucial to understand how these dormant cells are formed, and how sporulation can be prevented or disrupted with the view of reducing CDI. Here, we review the current literature on the DMP and Q:AH machineries in C. difficile, and how they compare and contrast to those of B. subtilis. Overview of the DMP and Q:AH engulfment machineries in C. difficile. Analyses of the conservation of DMP across Bacilli, Clostridia and other bacteria. Proposes a multi-protein complex required for engulfment: the engulfasome. Highlights differential arrangements of engulfasome in B. subtilis and C. difficile.
Collapse
Affiliation(s)
- Abigail Kelly
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Paula S Salgado
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
19
|
U.S.-Based National Surveillance for Fidaxomicin Susceptibility of Clostridioides difficile-Associated Diarrheal Isolates from 2013 to 2016. Antimicrob Agents Chemother 2019; 63:AAC.00391-19. [PMID: 31085514 DOI: 10.1128/aac.00391-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/06/2019] [Indexed: 12/26/2022] Open
Abstract
In 2011, we initiated a sentinel surveillance network to assess changes in Clostridioides (formerly Clostridium) difficile antimicrobial susceptibility to fidaxomicin from 6 geographically dispersed medical centers in the United States. This report summarizes data from 2013 to 2016. C. difficile isolates or toxin-positive stools from patients were referred to a central laboratory. Antimicrobial susceptibility was determined by agar dilution. CLSI, EUCAST, or FDA breakpoints were used, where applicable. Toxin gene profiles were characterized by multiplex PCR on each isolate. A random sample of approximately 40% of isolates, stratified by institution and year, was typed by restriction endonuclease analysis (REA). Among 1,889 isolates from 2013 to 2016, the fidaxomicin MIC90 was 0.5 μg/ml; all isolates were inhibited at ≤1 μg/ml. There were decreases in metronidazole and vancomycin MICs over time. Clindamycin resistance remained unchanged (27.3%). An increase in imipenem resistance was observed. By 2015 to 2016, moxifloxacin resistance decreased in all centers. The proportion of BI isolates decreased from 25.5% in 2011 to 2012 to 12.8% in 2015 to 2016 (P < 0.001). The BI REA group correlated with moxifloxacin resistance (BI 84% resistant versus non-BI 12.5% resistant). Fidaxomicin MICs have not changed among C. difficile isolates of U.S. origin over 5 years post licensure. There has been an overall decrease in MICs for vancomycin, metronidazole, moxifloxacin, and rifampin and an increase in isolates resistant to imipenem. Moxifloxacin resistance remained high among the BI REA group, but the proportion of BI isolates has decreased. Continued geographic variations in REA groups and antimicrobial resistance persist.
Collapse
|
20
|
McSweeney B, Allegretti JR, Fischer M, Xu H, Goodman KJ, Monaghan T, McLeod C, Mullish BH, Petrof EO, Phelps EL, Chis R, Edmison A, Juby A, Ennis-Davis R, Roach B, Wong K, Kao D. In search of stool donors: a multicenter study of prior knowledge, perceptions, motivators, and deterrents among potential donors for fecal microbiota transplantation. Gut Microbes 2019; 11:51-62. [PMID: 31122134 PMCID: PMC6973337 DOI: 10.1080/19490976.2019.1611153] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Fecal microbiota transplantation (FMT) is a highly effective therapy for recurrent Clostridioides difficile infection. Stool donors are essential, but difficult to recruit and retain. We aimed to identify factors influencing willingness to donate stool. This multi-center study with a 32-item questionnaire targeted young adults and health care workers via social media and university email lists in Edmonton and Kingston, Canada; London and Nottingham, England; and Indianapolis and Boston, USA. Items included baseline demographics and FMT knowledge and perception. Investigated motivators and deterrents included economic compensation, screening process, time commitment, and stool donation logistics. Logistic regression and linear regression models estimated associations of study variables with self-assessed willingness to donate stool. 802 respondents completed our questionnaire: 387 (48.3%) age 21-30 years, 573 (71.4%) female, 323 (40%) health care workers. Country of residence, age and occupation were not associated with willingness to donate stool. Factors increasing willingness to donate were: already a blood donor (OR 1.64), male, altruism, economic benefit, knowledge of how FMT can help patients (OR 1.32), and positive attitudes towards FMT (OR 1.39). Factors decreasing willingness to donate were: stool collection unpleasant (OR 0.92), screening process invasive (OR 0.92), higher stool donation frequency, negative social perception of stool, and logistics of collection/transporting feces. We conclude that 1) blood donors and males are more willing to consider stool donation; 2) altruism, economic compensation, and positive feedback are motivators; and 3) screening process, high donation frequency, logistics of collection/transporting feces, lack of public awareness, and negative social perception are deterrents. Considering these variables could maximize donor recruitment and retention.
Collapse
Affiliation(s)
- Breanna McSweeney
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Jessica R. Allegretti
- Division of Gastroenterology, Brigham and Women’s Hospital/Harvard Medical School, Boston, MA, USA
| | - Monika Fischer
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, IN, USA
| | - Huiping Xu
- Department of Biostatistics, The Richard M. Fairbanks School of Public Health and School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Karen J. Goodman
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Tanya Monaghan
- NIHR Nottingham Biomedical Research Centre (BRC), Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Carmen McLeod
- Nottingham Synthetic Biology Research Centre, School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Benjamin H. Mullish
- Division of Integrative Systems Medicine and Digestive Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Elaine O. Petrof
- Department of Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Emmalee L. Phelps
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, IN, USA
| | - Roxana Chis
- Department of Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Abby Edmison
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Angela Juby
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ralph Ennis-Davis
- Department of Medicine, Alberta Health Services, Edmonton, Alberta, Canada
| | - Brandi Roach
- Zeidler Ledcor Center, Division of Gastroenterology, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Wong
- Zeidler Ledcor Center, Division of Gastroenterology, University of Alberta, Edmonton, Alberta, Canada
| | - Dina Kao
- Zeidler Ledcor Center, Division of Gastroenterology, University of Alberta, Edmonton, Alberta, Canada,CONTACT Dina Kao Zeidler Ledcor Center, Division of Gastroenterology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Rubio-Terrés C, Aguado JM, Almirante B, Cobo J, Grau S, Salavert M, González Antona Sánchez E, López Gutiérrez C, Rubio-Rodríguez D. Extended-pulsed fidaxomicin versus vancomycin in patients 60 years and older with Clostridium difficile infection: cost-effectiveness analysis in Spain. Eur J Clin Microbiol Infect Dis 2019; 38:1105-1111. [PMID: 30989419 PMCID: PMC6520320 DOI: 10.1007/s10096-019-03503-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/04/2019] [Indexed: 01/13/2023]
Abstract
The cost of treating Clostridium difficile infection (CDI) in Spain is substantial. Findings from the randomised, controlled, open-label, phase 3b/4 EXTEND study showed that an extended-pulsed fidaxomicin (EPFX) regimen was associated with improved sustained clinical cure and reduced recurrence of CDI versus vancomycin in patients aged 60 years and older. We assessed the cost-effectiveness of EPFX versus vancomycin for the treatment of CDI in patients aged 60 years and older from the perspective of the National Health System (NHS) in Spain. We used a Markov model with six health states and 1-year time horizon. Health resources, their unit costs and utilities were based on published sources. Key efficacy data and transition probabilities were obtained from the EXTEND study and published sources. A panel of Spanish clinical experts validated all model assumptions. In the analysis, 0.638 and 0.594 quality-adjusted life years (QALYs) per patient were obtained with EPFX and vancomycin, respectively, with a gain of 0.044 QALYs with EPFX. The cost per patient treated with EPFX and vancomycin was estimated to be €10,046 and €10,693, respectively, with a saving of €647 per patient treated with EPFX. For willingness-to-pay thresholds of €20,000, €25,000 and €30,000 per QALY gained, the probability that EPFX was the most cost-effective treatment was 99.3%, 99.5% and 99.9%, respectively. According to our economic model and the assumptions based on the Spanish NHS, EPFX is cost-effective compared with vancomycin for the first-line treatment of CDI in patients aged 60 years and older.
Collapse
Affiliation(s)
| | - José María Aguado
- Department of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Benito Almirante
- Department of Infectious Diseases, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Javier Cobo
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal/IRYCIS, Madrid, Spain
| | - Santiago Grau
- Department of Pharmacy, Hospital del Mar, Barcelona, Spain
| | - Miguel Salavert
- Department of Infectious Diseases, Hospital Universitario La Fe, Valencia, Spain
| | | | | | | |
Collapse
|
22
|
Barbanti F, Spigaglia P. Direct detection and characterization of Clostridium difficile from a novel collection device to improve laboratory workflow. APMIS 2019; 127:449-453. [PMID: 30834561 DOI: 10.1111/apm.12942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 02/18/2019] [Indexed: 11/30/2022]
Abstract
Emergence of Clostridium difficile strains with increased virulence emphasizes the importance of early diagnosis and surveillance of C. difficile infection (CDI). In this study, the new FecalSwab™ collection and transport system was evaluated to improve C. difficile diagnosis. The FecalSwab™ was used for direct C. difficile molecular detection, C. difficile culture/toxigenic culture (TC) and bacterial genomic DNA (bgDNA) extraction. Our results demonstrated that the FecalSwab™ medium could be successfully used as template for Xpert C. difficile binary toxin (BT), regardless of the bacterial load of samples, and for C. difficile culture also after a long storage (30 days) of FecalSwab™ tubes at 4 °C. Furthermore, good-quality bgDNA was extracted from the FecalSwab™ medium for the majority (75%) of the samples analyzed. Typing was performed to fully characterize C. difficile strains isolated during this study and 17 different PCR-ribotypes (RTs) were identified. The results obtained indicate that the FecalSwab™ can be successfully used not only in daily diagnostic routine of C. difficile but also in surveillance and retrospective studies.
Collapse
Affiliation(s)
- Fabrizio Barbanti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Patrizia Spigaglia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
23
|
Peng Z, Wang S, Gide M, Zhu D, Lamabadu Warnakulasuriya Patabendige HM, Li C, Cai J, Sun X. A Novel Bacteriophage Lysin-Human Defensin Fusion Protein Is Effective in Treatment of Clostridioides difficile Infection in Mice. Front Microbiol 2019; 9:3234. [PMID: 30687250 PMCID: PMC6336692 DOI: 10.3389/fmicb.2018.03234] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/12/2018] [Indexed: 12/17/2022] Open
Abstract
Clostridioides difficile is the leading cause of worldwide antibiotics-associated diarrhea. In this study, we report the construction and evaluation of a novel bacteriophage lysin-human defensin fusion protein targeting C. difficile. The fusion protein, designated LHD, is composed of two parts connected by a 3-repeating unit linker "(GGGGS)3": the catalytic domain of a lysin protein from a C. difficile bacteriophage phiC2 (LCD), and the functional domain of a human defensin protein HD5. Lytic assays showed that LHD protein had a potent lytic activity against different types of clinical C. difficile strains, including the epidemic 027, 078, 012, and 087 strains. The minimum inhibitory concentration (MIC) of LHD was 0.78 μg/ml, which was lower than the MIC of the protein LCD (1.56 μg/ml), and the MICs of metronidazole (4 μg/ml) and vancomycin (4 μg/ml). In addition, the LHD protein could lyse C. different strains in different pHs (6.0, 7.0, and 8.0). Evaluation of LHD potency in vivo using mouse model of C. difficile infection (CDI) showed that administration of the LHD protein (twice daily for 7 days) was effective in mitigating the symptoms and reducing the death from CDI. Treatment with LHD also significantly decreased the number of C. difficile spores and the toxin level in feces from the infected mice. Our data suggest that this novel lysin-human defensin fusion protein has a potential on CDI control.
Collapse
Affiliation(s)
- Zhong Peng
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mussie Gide
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Duolong Zhu
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | | | - Chunhui Li
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
24
|
Salavert M, Cobo J, Pascual Á, Aragón B, Maratia S, Jiang Y, Aceituno S, Grau S. Cost-Effectiveness Analysis of Bezlotoxumab Added to Standard of Care Versus Standard of Care Alone for the Prevention of Recurrent Clostridium difficile Infection in High-Risk Patients in Spain. Adv Ther 2018; 35:1920-1934. [PMID: 30328061 PMCID: PMC6223985 DOI: 10.1007/s12325-018-0813-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Clostridium difficile infection (CDI) is the major cause of infectious nosocomial diarrhoea and is associated with considerable morbidity, mortality and economic impact. Bezlotoxumab administered in combination with standard of care (SoC) antibiotic therapy prevents recurrent CDI. This study assessed the cost-effectiveness of bezlotoxumab added to SoC, compared to SoC alone, to prevent the recurrence of CDI in high-risk patients from the Spanish National Health System perspective. METHODS A Markov model was used to simulate the natural history of CDI over a lifetime horizon in five populations of patients at high risk of CDI recurrence according to MODIFY trials: (1) ≥ 65 years old; (2) severe CDI; (3) immunocompromised; (4) ≥ 1 CDI episode in the previous 6 months; and (5) ≥ 65 years old and with ≥ 1 CDI episode in the previous 6 months. The incremental cost-effectiveness ratio (ICER) expressed as cost per quality-adjusted life-year (QALY) gained was calculated. Deterministic (DSA) and probabilistic sensitivity analyses (PSA) were performed. RESULTS In all patient populations (from 1 to 5), bezlotoxumab added to SoC reduced CDI recurrence compared to SoC alone by 26.4, 19.5, 21.2, 26.6 and 39.7%, respectively. The resulting ICERs for the respective subgroups were €12,724, €17,495, €9545, €7386, and €4378. The model parameters with highest impact on the ICER were recurrence rate (first), mortality, and utility values. The probability that bezlotoxumab was cost-effective at a willingness-to-pay threshold of €21,000/QALY was 85.5%, 54.1%, 86.0%, 94.5%, 99.6%, respectively. CONCLUSION The results suggest that bezlotoxumab added to SoC compared to SoC alone is a cost-effective treatment to prevent the recurrence of CDI in high-risk patients. The influence of changes in model parameters on DSA results was higher in patients ≥ 65 years old, with severe CDI and immunocompromised. Additionally, PSA estimated that the probability of cost-effectiveness exceeded 85% in most subgroups. FUNDING Merck Sharp & Dohme Corp.
Collapse
Affiliation(s)
- Miguel Salavert
- Infectious Diseases Unit, Hospital Universitario La Fe, Valencia, Spain.
| | - Javier Cobo
- Infectious Diseases Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Álvaro Pascual
- Clinical Microbiology and Infectious Diseases Unit, Hospital Universitario Virgen Macarena, Instituto de Biomedicina de Sevilla (IBIS), Universidad de Sevilla, Seville, Spain
| | | | | | - Yiling Jiang
- Merck Sharp & Dohme Ltd, Hoddesdon, Hertfordshire, UK
| | | | - Santiago Grau
- Pharmacy Department, Hospital del Mar, Universidad Autónoma de Barcelona, Barcelona, Spain
| |
Collapse
|
25
|
Bernal I, Hofmann JD, Bulitta B, Klawonn F, Michel AM, Jahn D, Neumann-Schaal M, Bruder D, Jänsch L. Clostridioides difficile Activates Human Mucosal-Associated Invariant T Cells. Front Microbiol 2018; 9:2532. [PMID: 30410474 PMCID: PMC6209678 DOI: 10.3389/fmicb.2018.02532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022] Open
Abstract
Clostridioides difficile infection (CDI) causes severe inflammatory responses at the intestinal mucosa but the immunological mechanisms underlying CDI-related immunopathology are still incompletely characterized. Here we identified for the first time that both, non-toxigenic strains as well as the hypervirulent ribotypes RT027 and RT023 of Clostridioides difficile (formerly Clostridium difficile), induced an effector phenotype in mucosal-associated invariant T (MAIT) cells. MAIT cells can directly respond to bacterial infections by recognizing MR1-presented metabolites derived from the riboflavin synthesis pathway constituting a novel class of antigens. We confirmed functional riboflavin synthesis of C. difficile and found fixed bacteria capable of activating primary human MAIT cells in a dose-dependent manner. C. difficile-activated MAIT cells showed an increased and MR1-dependent expression of CD69, proinflammatory IFNγ, and the lytic granule components granzyme B and perforin. Effector protein expression was accompanied by the release of lytic granules, which, in contrast to other effector functions, was mainly induced by IL-12 and IL-18. Notably, this study revealed hypervirulent C. difficile strains to be most competent in provoking MAIT cell responses suggesting MAIT cell activation to be instrumental for the immunopathology observed in C. difficile-associated colitis. In conclusion, we provide first evidence for a link between C. difficile metabolism and innate T cell-mediated immunity in humans.
Collapse
Affiliation(s)
- Isabel Bernal
- Institute of Medical Microbiology and Hospital Hygiene, Infection Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany.,ESF Graduate School ABINEP, Magdeburg, Germany
| | - Julia Danielle Hofmann
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | | | - Frank Klawonn
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Computer Science, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany
| | - Annika-Marisa Michel
- Department of Microbiology, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Dieter Jahn
- Department of Microbiology, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Meina Neumann-Schaal
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany.,Leibniz Institute DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Dunja Bruder
- Institute of Medical Microbiology and Hospital Hygiene, Infection Immunology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|