51
|
Meryk A, Kropshofer G, Bargehr C, Knoll M, Hetzer B, Lass-Flörl C, Crazzolara R. Which Type of Empiric Antibiotic Therapy is Appropriate? A 20-Year Retrospective Study of Bloodstream Infections in Childhood Cancer. Infect Dis Ther 2021; 10:789-800. [PMID: 33704685 PMCID: PMC8116419 DOI: 10.1007/s40121-021-00427-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Sufficient empirical antimicrobial therapy in febrile patients with cancer is challenging, owing to the limited arsenal of available antibiotics in an era of growing resistance. Because of the emergence of gram-negative bacteria resistant to ceftazidime and piperacillin, a combination antibiotic therapy was employed that uses meropenem combined with gentamicin and/or vancomycin if the patient further deteriorates. METHODS A retrospective cohort analysis was performed including all patients with catheter-associated bloodstream infections (BSIs) and treated for childhood cancer in a tertiary single centre between 1 January 2000 and 31 June 2018. We calculated the prevalence and the risk for BSIs and compared the in vitro susceptibility to various antimicrobial agents. RESULTS Of 653 patients with childhood cancer, 113 patients (17.3%) were identified with a total of 139 BSIs, most of them occurring in patients with leukaemia (n = 90, 64.7%) and were associated with gram-positive bacteria (60.5%). In our cohort, all BSIs with gram-negative bacteria exhibited in vitro susceptibility against meropenem alone without any signs of resistance development. The antibiotic coverage of our meropenem-based combination therapy was also highly effective for gram-positive and non-fermenting bacteria. Thus, BSI-related mortality in all 139 BSI episodes was 1.4%. Clostridium difficile infections (CDIs), as main adverse event of carbapenem usage, occurred in only 16 (2.5%) patients. CONCLUSION Our meropenem-based combination therapy showed sufficient empirical antibiotic coverage in the majority of BSIs (96.4%) and did not result in an increased rate of unwanted side effects or development of antibiotic resistance.
Collapse
Affiliation(s)
- Andreas Meryk
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Kropshofer
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Caroline Bargehr
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Miriam Knoll
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Benjamin Hetzer
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Roman Crazzolara
- Department of Pediatrics, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
52
|
Engineered Phage Endolysin Eliminates Gardnerella Biofilm without Damaging Beneficial Bacteria in Bacterial Vaginosis Ex Vivo. Pathogens 2021; 10:pathogens10010054. [PMID: 33435575 PMCID: PMC7830407 DOI: 10.3390/pathogens10010054] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
Bacterial vaginosis is characterized by an imbalance of the vaginal microbiome and a characteristic biofilm formed on the vaginal epithelium, which is initiated and dominated by Gardnerella bacteria, and is frequently refractory to antibiotic treatment. We investigated endolysins of the type 1,4-beta-N-acetylmuramidase encoded on Gardnerella prophages as an alternative treatment. When recombinantly expressed, these proteins demonstrated strong bactericidal activity against four different Gardnerella species. By domain shuffling, we generated several engineered endolysins with 10-fold higher bactericidal activity than any wild-type enzyme. When tested against a panel of 20 Gardnerella strains, the most active endolysin, called PM-477, showed minimum inhibitory concentrations of 0.13–8 µg/mL. PM-477 had no effect on beneficial lactobacilli or other species of vaginal bacteria. Furthermore, the efficacy of PM-477 was tested by fluorescence in situ hybridization on vaginal samples of fifteen patients with either first time or recurring bacterial vaginosis. In thirteen cases, PM-477 killed the Gardnerella bacteria and physically dissolved the biofilms without affecting the remaining vaginal microbiome. The high selectivity and effectiveness in eliminating Gardnerella, both in cultures of isolated strains as well as in clinically derived samples of natural polymicrobial biofilms, makes PM-477 a promising alternative to antibiotics for the treatment of bacterial vaginosis, especially in patients with frequent recurrence.
Collapse
|
53
|
Knežević D, Petković M. Faecal transplantation and Clostridioides difficile infection. SCRIPTA MEDICA 2021. [DOI: 10.5937/scriptamed52-32752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Faecal microbiota transplantation (FMT), known equally well as faecal transplantation or faecal bacteriotherapy, is the process of implanting the faecal suspension containing balanced microbiota from a healthy donor to the colon of a recipient patient. Excessive growth of Clostridioides difficile (C difficile) in the intestinal microbiota resulting from antibiotic consumption is currently a rising threat to public health. FMT is one of the most important, newer approaches to treating C difficile infections. Since C difficile is regarded as an opportunistic bacterium triggering disease in conditions of disturbed homeostasis of the intestinal microbiota, restoration of healthy intestinal microflora facilitates suppression of toxic strain of C difficile by anaerobic bacteria of normal intestinal microflora with concomitant cure. Nurses have important role in caring for patients after faecal transplantation.
Collapse
|
54
|
Gu SL, Gong Y, Zhang J, Chen Y, Wu Z, Xu Q, Fang Y, Wang J, Tang LL. Effect of the Short-Term Use of Fluoroquinolone and β-Lactam Antibiotics on Mouse Gut Microbiota. Infect Drug Resist 2020; 13:4547-4558. [PMID: 33376361 PMCID: PMC7762438 DOI: 10.2147/idr.s281274] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Antibiotics play an important role in the treatment of infectious diseases. However, the overuse of antibiotics increases the spread of drug-resistant bacteria and causes dysbiosis of the intestinal microbiota. Few studies have addressed the longitudinal effects of antibiotics on the microbiome and host immunity. MATERIALS AND METHODS In this study, the short-term effect of fluoroquinolone (levofloxacin) and β-lactam antibiotics (meropenem, cefoperazone/sulbactam, and aztreonam) on the gut microbiota of mice was evaluated by 16S rRNA gene sequencing. The susceptibility of Bifidobacterium longum, Lactobacillus lactis, Enterococcus faecium, and Clostridium butyricum to these antimicrobial agents was assessed using the disc diffusion method. RESULTS Our results showed that 4-day antibiotic exposure significantly reduced the alpha and beta diversity of gut bacteria and increased serum inflammatory cytokines, and these changes persisted long after antibiotic withdrawal and did not return to pre-treatment levels. Nonetheless, the bacterial community composition tended to return to pre-treatment levels after discontinuing treatment. The tested probiotic strains were resistant to aztreonam but were sensitive to meropenem and cefoperazone/sulbactam. CONCLUSION Short-term antibiotic treatment led to significant changes in the intestinal flora with a tendency to recover. The antibiotics had different effects on the intestinal microbial community and probiotic strains. This study provides guidance for the concomitant use of probiotics and antibiotics, and the results emphasize the importance of using broad-spectrum antibiotics responsibly to prevent the long-term disruption of the native microbiota.
Collapse
Affiliation(s)
- Si-Lan Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Yiwen Gong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Jiaying Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Zhengjie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Qiaomai Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Yunhui Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Jingxia Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, People’s Republic of China
| | - Ling-Ling Tang
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Shulan International Medical College, Hangzhou310000, People’s Republic of China
| |
Collapse
|
55
|
Vedamurthy A, Rajendran I, Manian F. Things We Do for No Reason™: Routine Coverage of Anaerobes in Aspiration Pneumonia. J Hosp Med 2020; 15:754-756. [PMID: 32966204 DOI: 10.12788/jhm.3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 03/25/2020] [Indexed: 11/20/2022]
Affiliation(s)
- Amar Vedamurthy
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Iniya Rajendran
- Department of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Farrin Manian
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
56
|
Patel B, Noda A, Godbout E, Stevens M, Noda C. Levofloxacin for Antibacterial Prophylaxis in Pediatric Patients With Acute Myeloid Leukemia or Undergoing Hematopoietic Stem Cell Transplantation. J Pediatr Pharmacol Ther 2020; 25:629-635. [PMID: 33041718 DOI: 10.5863/1551-6776-25.7.629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE This study aimed to evaluate the use of levofloxacin for the prevention of bacterial infections in pediatric patients with acute myeloid leukemia or those undergoing hematopoietic stem cell transplantation. METHODS This study was a single-center, retrospective review designed to assess the frequency of bacteremia with levofloxacin prophylaxis compared with historical controls that used other, clinician-directed antibacterial prophylaxis. The primary outcome of the study was microbiologically documented bacteremia. Secondary outcomes included febrile neutropenia, clinically documented infection, duration of neutropenia, treatment antibiotic exposure days, Clostridioides difficile infection, and infection-related mortality. RESULTS Of the 60 patients included, 24 patients with 32 hospital admissions received levofloxacin and 36 patients with 48 hospital admissions received clinician-directed prophylaxis. There was no difference found in the frequency of bacteremia between levofloxacin and clinician-directed prophylaxis (15.6% vs 10.4%, p = 0.49). There was no difference in the incidence of febrile neutropenia, clinically documented infection, treatment antibiotic exposure days, or 30-day infection-related mortality between the 2 groups. The levofloxacin group had a longer mean duration of neutropenia compared with clinician-directed prophylaxis (26.8 days vs 16.4 days, p = 0.01). CONCLUSIONS There was no difference in bacteremia between levofloxacin prophylaxis and clinician-directed prophylaxis in pediatric patients with acute myeloid leukemia or those undergoing hematopoietic stem cell transplantation. Levofloxacin prophylaxis is an appropriate alternative for the prevention of serious bacterial infections in this patient population, although further studies are needed to confirm these results.
Collapse
|
57
|
Tischendorf J, Brunner M, Knobloch MJ, Schulz L, Barker A, Wright MO, Lepak A, Safdar N. Evaluation of a successful fluoroquinolone restriction intervention among high-risk patients: A mixed-methods study. PLoS One 2020; 15:e0237987. [PMID: 32841259 PMCID: PMC7446965 DOI: 10.1371/journal.pone.0237987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/06/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE We conducted a quality improvement initiative to restrict fluoroquinolone prescribing on two inpatient units housing high-risk patients and applied a human factors approach to understanding the barriers and facilitators to success of this intervention by front-line providers. METHODS This was a mixed-methods, quasi-experimental study. This study was conducted on two inpatient units at a tertiary care academic medical center: the medical-surgical intensive care and abdominal solid organ transplant units. Unit-level data were collected retrospectively for 24 months pre- and post- fluoroquinolone restriction intervention, implemented in July 2016, for all admissions to the study units. Our restriction intervention required antimicrobial stewardship pre-approval for fluoroquinolone prescribing. We explored barriers and facilitators to optimal fluoroquinolone prescribing using semi-structured interviews attending, fellow and resident physicians, advanced practice providers and pharmacists on these units. RESULTS Hospital-onset C. difficile infection did not decrease significantly, but fluoroquinolone use declined significantly from 111.6 to 19.8 days of therapy per 1000 patient-days without negatively impacting length of stay, readmissions or mortality. Third generation cephalosporin and aminoglycoside use increased post-restriction. Providers identified our institution's strong antimicrobial stewardship program and pharmacy involvement in antimicrobial decision making as key facilitators of fluoroquinolone optimization and patient complexity, lack of provider education and organizational culture as barriers to optimal prescribing. CONCLUSIONS Fluoroquinolones can be safely restricted even among high-risk patients without negatively impacting length of stay, readmissions or mortality. Our study provides a framework for successful antimicrobial stewardship interventions informed by perceptions of front line providers.
Collapse
Affiliation(s)
- Jessica Tischendorf
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Matthew Brunner
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Mary Jo Knobloch
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| | - Lucas Schulz
- Department of Pharmacy, University of Wisconsin Hospital and Clinics, Madison, Wisconsin, United States of America
| | - Anna Barker
- Department of Population Health Sciences, University of Wisconsin Madison, Madison, Wisconsin, United States of America
- Department of Internal Medicine, University of Michigan Ann Arbor, Ann Arbor, Michigan, United States of America
| | - Marc-Oliver Wright
- Infection Prevention Program, University of Wisconsin Hospital and Clinics, Madison, Wisconsin, United States of America
| | - Alexander Lepak
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Nasia Safdar
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| |
Collapse
|
58
|
Identification of Simplified Microbial Communities That Inhibit Clostridioides difficile Infection through Dilution/Extinction. mSphere 2020; 5:5/4/e00387-20. [PMID: 32727857 PMCID: PMC7392540 DOI: 10.1128/msphere.00387-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal microbiome plays an important role in limiting susceptibility to infection with Clostridioides difficile To better understand the ecology of bacteria important for C. difficile colonization resistance, we developed an experimental platform to simplify complex communities of fecal bacteria through dilution and rapidly screen for their ability to resist C. difficile colonization after challenge, as measured by >100-fold reduction in levels of C. difficile in challenged communities. We screened 76 simplified communities diluted from cultures of six fecal donors and identified 24 simplified communities that inhibited C. difficile colonization in vitro Sequencing revealed that simplified communities were composed of 19 to 67 operational taxonomic units (OTUs) and could be partitioned into four distinct community types. One simplified community could be further simplified from 56 to 28 OTUs through dilution and retain the ability to inhibit C. difficile We tested the efficacy of seven simplified communities in a humanized microbiota mouse model. We found that four communities were able to significantly reduce the severity of the initial C. difficile infection and limit susceptibility to disease relapse. Analysis of fecal microbiomes from treated mice demonstrated that simplified communities accelerated recovery of indigenous bacteria and led to stable engraftment of 19 to 22 OTUs from simplified communities. Overall, the insights gained through the identification and characterization of these simplified communities increase our understanding of the microbial dynamics of C. difficile infection and recovery.IMPORTANCEClostridioides difficile is the leading cause of antibiotic-associated diarrhea and a significant health care burden. Fecal microbiota transplantation is highly effective at treating recurrent C. difficile disease; however, uncertainties about the undefined composition of fecal material and potential long-term unintended health consequences remain. These concerns have motivated studies to identify new communities of microbes with a simpler composition that will be effective at treating disease. This work describes a platform for rapidly identifying and screening new simplified communities for efficacy in treating C. difficile infection. Four new simplified communities of microbes with potential for development of new therapies to treat C. difficile disease are identified. While this platform was developed and validated to model infection with C. difficile, the underlying principles described in the paper could be easily modified to develop therapeutics to treat other gastrointestinal diseases.
Collapse
|
59
|
Bhalodi AA, van Engelen TSR, Virk HS, Wiersinga WJ. Impact of antimicrobial therapy on the gut microbiome. J Antimicrob Chemother 2020; 74:i6-i15. [PMID: 30690540 PMCID: PMC6382031 DOI: 10.1093/jac/dky530] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The gut microbiome is now considered an organ unto itself and plays an important role in health maintenance and recovery from critical illness. The commensal organisms responsible for the framework of the gut microbiome are valuable in protection against disease and various physiological tasks. Critical illness and the associated interventions have a detrimental impact on the microbiome. While antimicrobials are one of the fundamental and often life-saving modalities in septic patients, they can also pave the way for subsequent harm because of the resulting damage to the gut microbiome. Contributing to many of the non-specific signs and symptoms of sepsis, the balance between the overuse of antimicrobials and the clinical need in these situations is often difficult to delineate. Given the potency of antimicrobials utilized to treat septic patients, the effects on the gut microbiome are often rapid and long-lasting, in which case full recovery may never be observed. The overgrowth of opportunistic pathogens is of significant concern as they can lead to infections that become increasingly difficult to treat. Continued research to understand the disturbances within the gut microbiome of critically ill patients and their outcomes is essential to help develop future therapies to circumvent damage to, or restore, the microbiome. In this review, we discuss the impact of the antimicrobials often used for the treatment of sepsis on the gut microbiota.
Collapse
Affiliation(s)
- Amira A Bhalodi
- Accelerate Diagnostics, Inc., Scientific Affairs, Tucson, AZ, USA
| | - Tjitske S R van Engelen
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - Harjeet S Virk
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - W Joost Wiersinga
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands.,Amsterdam UMC, University of Amsterdam, Department of Medicine, Division of Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
60
|
Rhea S, Jones K, Endres-Dighe S, Munoz B, Weber DJ, Hilscher R, MacFarquhar J, Sickbert-Bennett E, DiBiase L, Marx A, Rineer J, Lewis J, Bobashev G. Modeling inpatient and outpatient antibiotic stewardship interventions to reduce the burden of Clostridioides difficile infection in a regional healthcare network. PLoS One 2020; 15:e0234031. [PMID: 32525887 PMCID: PMC7289388 DOI: 10.1371/journal.pone.0234031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 05/19/2020] [Indexed: 12/14/2022] Open
Abstract
Antibiotic exposure can lead to unintended outcomes, including drug-drug interactions, adverse drug events, and healthcare-associated infections like Clostridioides difficile infection (CDI). Improving antibiotic use is critical to reduce an individual's CDI risk. Antibiotic stewardship initiatives can reduce inappropriate antibiotic prescribing (e.g., unnecessary antibiotic prescribing, inappropriate antibiotic selection), impacting both hospital (healthcare)-onset (HO)-CDI and community-associated (CA)-CDI. Previous computational and mathematical modeling studies have demonstrated a reduction in CDI incidence associated with antibiotic stewardship initiatives in hospital settings. Although the impact of antibiotic stewardship initiatives in long-term care facilities (LTCFs), including nursing homes, and in outpatient settings have been documented, the effects of specific interventions on CDI incidence are not well understood. We examined the relative effectiveness of antibiotic stewardship interventions on CDI incidence using a geospatially explicit agent-based model of a regional healthcare network in North Carolina. We simulated reductions in unnecessary antibiotic prescribing and inappropriate antibiotic selection with intervention scenarios at individual and network healthcare facilities, including short-term acute care hospitals (STACHs), nursing homes, and outpatient locations. Modeled antibiotic prescription rates were calculated using patient-level data on antibiotic length of therapy for the 10 modeled network STACHs. By simulating a 30% reduction in antibiotics prescribed across all inpatient and outpatient locations, we found the greatest reductions on network CDI incidence among tested scenarios, namely a 17% decrease in HO-CDI incidence and 7% decrease in CA-CDI. Among intervention scenarios of reducing inappropriate antibiotic selection, we found a greater impact on network CDI incidence when modeling this reduction in nursing homes alone compared to the same intervention in STACHs alone. These results support the potential importance of LTCF and outpatient antibiotic stewardship efforts on network CDI burden and add to the evidence that a coordinated approach to antibiotic stewardship across multiple facilities, including inpatient and outpatient settings, within a regional healthcare network could be an effective strategy to reduce network CDI burden.
Collapse
Affiliation(s)
- Sarah Rhea
- RTI International, Research Triangle Park, North Carolina
- * E-mail:
| | - Kasey Jones
- RTI International, Research Triangle Park, North Carolina
| | | | - Breda Munoz
- RTI International, Research Triangle Park, North Carolina
| | | | | | - Jennifer MacFarquhar
- North Carolina Department of Health and Human Services, Raleigh, North Carolina
- Career Epidemiology Field Officer Program, Division of State and Local Readiness, Center for Preparedness and Response, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | - Ashley Marx
- UNC Health Care, Chapel Hill, North Carolina
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina
| | - James Rineer
- RTI International, Research Triangle Park, North Carolina
| | - James Lewis
- North Carolina Department of Health and Human Services, Raleigh, North Carolina
| | | | | |
Collapse
|
61
|
Clostridium difficile Infection Epidemiology over a Period of 8 Years—A Single Centre Study. SUSTAINABILITY 2020. [DOI: 10.3390/su12114439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Clostridium difficile infection (CDI) is the most common infectious disease related to antibiotic-associated diarrhoea and is a current leading cause of morbidity/mortality, with substantial consequences for healthcare services and overall public health. Thus, we performed a retrospective epidemiological study of CDI for a long period (8 years), in an infectious hospital located in north-western Romania, which serves an entire county of the country (617,827 inhabitants). From 2011 to 2018, 877 patients were diagnosed with CDI; the mean incidence of this disease was 2.76 cases/10,000 patient-days, with an increasing trend in the annual incidence until 2016, at which point there was a decrease. The most commonly afflicted were patients in the 75–84 age group, observed in winter and spring. The results show that the antibiotics were administered in 679 (77.42%) subjects, within the last 3 months before CDI, statistically significant more than proton-pump inhibitors (PPIs)—128 (14.60%) and antidepressant medications—60 (6.84%), which were administered during the same period (p < 0.001). No medication was reported in 10 (6.84%) cases of CDI, in the last 3 months of the study. The fatality rate attained 4.1%, tripling in 2018 vs. 2011. CDI became a significant public health conundrum that can, nevertheless, be combatted through a judicious use of antibiotics.
Collapse
|
62
|
Yuan Y, Wang X, Xu X, Liu Y, Li C, Yang M, Yang Y, Ma Z. Evaluation of a Dual-Acting Antibacterial Agent, TNP-2092, on Gut Microbiota and Potential Application in the Treatment of Gastrointestinal and Liver Disorders. ACS Infect Dis 2020; 6:820-831. [PMID: 31849218 DOI: 10.1021/acsinfecdis.9b00374] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
TNP-2092 is a unique multitargeting drug conjugate with extremely low propensity for development of resistance. The in vitro activity of TNP-2092 against a panel of urease-producing bacteria was similar to that of rifaximin, a locally acting antibiotic approved for the treatment of hepatic encephalopathy, irritable bowel syndrome with diarrhea, and traveler's diarrhea. When given orally, TNP-2092 exhibited low absorption and the majority of compound was recovered in feces as parent. The impact of oral TNP-2092 on gut microbiota was investigated in rats. TNP-2092 was administered to rats by oral gavage for 7 days. Feces samples were collected and analyzed by 16S rRNA sequencing. Although the total amount of bacterial load appeared relatively unchanged before, during, and after treatment, significant changes in the relative abundance of certain gut bacteria at family and genus levels were observed. Some of the changes are known to be associated with improvement of symptoms associated with liver cirrhosis and hepatic encephalopathy. The observed effects of TNP-2092 on gut microbiota in rats were similar to those of rifaximin. In vivo, TNP-2092 demonstrated potent efficacy in a mouse Clostridium difficile infection model, superior to metronidazole and vancomycin, with no relapse observed after treatment. TNP-2092 is currently in clinical development for the treatment of symptoms associated with gastrointestinal and liver disorders.
Collapse
Affiliation(s)
- Ying Yuan
- TenNor Therapeutics Limited, 218 Xinghu Street, Suzhou Industrial Park, Suzhou 215123, China
| | - Xiaomei Wang
- TenNor Therapeutics Limited, 218 Xinghu Street, Suzhou Industrial Park, Suzhou 215123, China
| | - Xiangyi Xu
- TenNor Therapeutics Limited, 218 Xinghu Street, Suzhou Industrial Park, Suzhou 215123, China
| | - Yu Liu
- TenNor Therapeutics Limited, 218 Xinghu Street, Suzhou Industrial Park, Suzhou 215123, China
| | - Cancan Li
- WuXi AppTec (Shanghai) Co., Ltd., 288 Fute Zhong Road, Waigaoqiao, China (Shanghai) Pilot Free Trade Zone, Shanghai 200131, China
| | - Meng Yang
- WuXi AppTec (Shanghai) Co., Ltd., 288 Fute Zhong Road, Waigaoqiao, China (Shanghai) Pilot Free Trade Zone, Shanghai 200131, China
| | - Yiqing Yang
- BGI Genomics, BGI Park, No.21 Hongan 3rd Street, Yantian District, Shenzhen 518083, China
| | - Zhenkun Ma
- TenNor Therapeutics Limited, 218 Xinghu Street, Suzhou Industrial Park, Suzhou 215123, China
| |
Collapse
|
63
|
Antibiotic Exposure and Risk for Hospital-Associated Clostridioides difficile Infection. Antimicrob Agents Chemother 2020; 64:AAC.02169-19. [PMID: 31964789 DOI: 10.1128/aac.02169-19] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/11/2020] [Indexed: 01/08/2023] Open
Abstract
Clostridioides difficile infection (CDI) is a health care-associated infection associated with significant morbidity and cost, with highly varied risk across populations. More effective, risk-based prevention strategies are needed. Here, we investigate risk factors for hospital-associated CDI in a large integrated health system. In a retrospective cohort of all adult admissions to 21 Intermountain Healthcare hospitals from 2006 to 2012, we identified all symptomatic (i) hospital-onset and (ii) health care-facility-associated, community-onset CDI. We then evaluated the risk associated with antibiotic exposure, including that of specific agents, using multivariable logistic regression. A total of 2,356 cases of CDI among 506,068 admissions were identified (incidence, 46.6 per 10,000). Prior antibiotic use was the dominant risk factor, where for every antibiotic day of therapy prior to the index admission, the odds of subsequent CDI increased by 12.8% (95% confidence interval [CI], 12.2 to 13.4%; P < 0.0001). This was a much stronger association than was inpatient antibiotic exposure (odds ratio [OR], 1.007 [95% CI, 1.005 to 1.009]; P < 0.0001). The highest-risk antibiotics included second-generation and later cephalosporins (especially oral), carbapenems, fluoroquinolones, and clindamycin, while doxycycline and daptomycin were associated with a lower CDI risk. We concluded that cumulative antibiotic exposure prior to admission is the greatest contributor to the risk of subsequent CDI. Most classes of antibiotics carry some risk, which varies by drug and route. This information may be useful for antimicrobial stewardship efforts.
Collapse
|
64
|
Fuzi M, Rodriguez Baño J, Toth A. Global Evolution of Pathogenic Bacteria With Extensive Use of Fluoroquinolone Agents. Front Microbiol 2020; 11:271. [PMID: 32158437 PMCID: PMC7052298 DOI: 10.3389/fmicb.2020.00271] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
It is well-established that the spread of many multidrug-resistant (MDR) bacteria is predominantly clonal. Interestingly the international clones/sequence types (STs) of most pathogens emerged and disseminated during the last three decades. Strong experimental evidence from multiple laboratories indicate that diverse fitness cost associated with high-level resistance to fluoroquinolones contributed to the selection and promotion of the international clones/STs of hospital-associated methicillin-resistant Staphylococcus aureus (HA-MRSA), extended-spectrum β-lactamase-(ESBL)-producing Klebsiella pneumoniae, ESBL-producing Escherichia coli and Clostridioides difficile. The overwhelming part of the literature investigating the epidemiology of the pathogens as a function of fluoroquinolone use remain in concordence with these findings. Moreover, recent in vitro data clearly show the potential of fluoroquinolone exposure to shape the clonal evolution of Salmonella Enteritidis. The success of the international clones/STs in all these species was linked to the strains' unique ability to evolve multiple energetically beneficial gyrase and topoisomerase IV mutations conferring high-level resistance to fluorquinolones and concomittantly permitting the acquisition of an extra resistance gene load without evoking appreciable fitness cost. Furthermore, by analyzing the clonality of multiple species, the review highlights, that in environments under high antibiotic exposure virulence factors play only a subsidiary role in the clonal dynamics of bacteria relative to multidrug-resistance coupled with favorable fitness (greater speed of replication). Though other groups of antibiotics should also be involved in selecting clones of bacterial pathogens the role of fluoroquinolones due to their peculiar fitness effect remains unique. It is suggested that probably no bacteria remain immune to the influence of fluoroquinolones in shaping their evolutionary dynamics. Consequently a more judicious use of fluoroquinolones, attuned to the proportion of international clone/ST isolates among local pathogens, would not only decrease resistance rates against this group of antibiotics but should also ameliorate the overall antibiotic resistance landscape.
Collapse
Affiliation(s)
- Miklos Fuzi
- Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary
| | - Jesus Rodriguez Baño
- Unit of Infectious Diseases, Clinical Microbiology and Preventive Medicine, Department of Medicine, Hospital Universitario Virgen Macarena, University of Seville - Biomedicine Institute of Seville (IBiS), Seville, Spain
| | - Akos Toth
- Department of Bacteriology, Mycology and Parasitology, National Public Health Center, Budapest, Hungary
| |
Collapse
|
65
|
SYN-007, an Orally Administered Beta-Lactamase Enzyme, Protects the Gut Microbiome from Oral Amoxicillin/Clavulanate without Adversely Affecting Antibiotic Systemic Absorption in Dogs. Microorganisms 2020; 8:microorganisms8020152. [PMID: 31979034 PMCID: PMC7074739 DOI: 10.3390/microorganisms8020152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/28/2022] Open
Abstract
Beta-lactamases, enzymes produced by bacteria to degrade beta-lactam antibiotics, have been harnessed as therapeutics to protect the gut microbiome from damage caused by antibiotics. Proof-of-concept of this approach using SYN-004 (ribaxamase), a beta-lactamase formulated for oral delivery with intravenous (IV) penicillins and cephalosporins, was demonstrated with animal models and in humans. Ribaxamase degraded ceftriaxone in the gastrointestinal tract, protected the gut microbiome, significantly reduced the incidence of Clostridioides difficile disease and attenuated emergence of antibiotic resistant organisms. SYN-007 is a delayed release formulation of ribaxamase intended for use with oral beta-lactams. In dogs treated with oral amoxicillin, SYN-007 diminished antibiotic-mediated microbiome disruption and reduced the emergence of antibiotic resistance without altering amoxicillin systemic absorption. Here, SYN-007 function in the presence of clavulanate, a beta-lactamase inhibitor, was investigated. Dogs received amoxicillin (40 mg/kg, orally (PO), three times a day (TID)) or the combined antibiotic/beta-lactamase inhibitor, amoxicillin/clavulanate (40 mg/kg amoxicillin, 5.7 mg/kg clavulanate, PO, TID) +/™ SYN-007 (10 mg, PO, TID) for five days. Serum amoxicillin levels were not significantly different +/™ SYN-007 compared to amoxicillin alone or amoxicillin/clavulanate alone as controls for both first and last doses, indicating SYN-007 did not interfere with systemic absorption of the antibiotic. Whole genome shotgun metagenomics analyses of the fecal microbiomes demonstrated both amoxicillin and amoxicillin/clavulanate significantly reduced diversity and increased the frequency of antibiotic resistance genes. Microbiome damage appeared more severe with amoxicillin/clavulanate. In contrast, with SYN-007, microbiome diversity was not significantly altered, and frequency of antibiotic resistance genes did not increase. Importantly, SYN-007 functioned in the presence of clavulanate to protect the gut microbiome indicating that SYN-007 activity was not inhibited by clavulanate in the dog gastrointestinal tract. SYN-007 has the potential to expand microbiome protection to beta-lactam/beta-lactamase inhibitor combinations delivered orally or systemically.
Collapse
|
66
|
Xu X, Godoy-Ruiz R, Adipietro KA, Peralta C, Ben-Hail D, Varney KM, Cook ME, Roth BM, Wilder PT, Cleveland T, Grishaev A, Neu HM, Michel SLJ, Yu W, Beckett D, Rustandi RR, Lancaster C, Loughney JW, Kristopeit A, Christanti S, Olson JW, MacKerell AD, Georges AD, Pozharski E, Weber DJ. Structure of the cell-binding component of the Clostridium difficile binary toxin reveals a di-heptamer macromolecular assembly. Proc Natl Acad Sci U S A 2020; 117:1049-1058. [PMID: 31896582 PMCID: PMC6969506 DOI: 10.1073/pnas.1919490117] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Targeting Clostridium difficile infection is challenging because treatment options are limited, and high recurrence rates are common. One reason for this is that hypervirulent C. difficile strains often have a binary toxin termed the C. difficile toxin, in addition to the enterotoxins TsdA and TsdB. The C. difficile toxin has an enzymatic component, termed CDTa, and a pore-forming or delivery subunit termed CDTb. CDTb was characterized here using a combination of single-particle cryoelectron microscopy, X-ray crystallography, NMR, and other biophysical methods. In the absence of CDTa, 2 di-heptamer structures for activated CDTb (1.0 MDa) were solved at atomic resolution, including a symmetric (SymCDTb; 3.14 Å) and an asymmetric form (AsymCDTb; 2.84 Å). Roles played by 2 receptor-binding domains of activated CDTb were of particular interest since the receptor-binding domain 1 lacks sequence homology to any other known toxin, and the receptor-binding domain 2 is completely absent in other well-studied heptameric toxins (i.e., anthrax). For AsymCDTb, a Ca2+ binding site was discovered in the first receptor-binding domain that is important for its stability, and the second receptor-binding domain was found to be critical for host cell toxicity and the di-heptamer fold for both forms of activated CDTb. Together, these studies represent a starting point for developing structure-based drug-design strategies to target the most severe strains of C. difficile.
Collapse
Affiliation(s)
- Xingjian Xu
- City University of New York Advanced Science Research Center, City University of New York, New York, NY 10017
- PhD Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10017
| | - Raquel Godoy-Ruiz
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Kaylin A Adipietro
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Christopher Peralta
- City University of New York Advanced Science Research Center, City University of New York, New York, NY 10017
| | - Danya Ben-Hail
- City University of New York Advanced Science Research Center, City University of New York, New York, NY 10017
| | - Kristen M Varney
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Mary E Cook
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Braden M Roth
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Paul T Wilder
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| | | | | | - Heather M Neu
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, MD 21201
| | - Sarah L J Michel
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, MD 21201
| | - Wenbo Yu
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, MD 21201
| | - Dorothy Beckett
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD 20742
| | | | | | | | | | | | | | - Alexander D MacKerell
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
- University of Maryland School of Pharmacy, University of Maryland, Baltimore, MD 21201
| | - Amedee des Georges
- City University of New York Advanced Science Research Center, City University of New York, New York, NY 10017;
- PhD Program in Biochemistry, The Graduate Center, City University of New York, New York, NY 10017
- PhD Program in Chemistry, The Graduate Center, City University of New York, New York, NY 10017
- Department of Chemistry & Biochemistry, City College of New York, New York, NY 10031
| | - Edwin Pozharski
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201;
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| | - David J Weber
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201;
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850
- The Center for Biomolecular Therapeutics, The University of Maryland School of Medicine, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
67
|
Ortigão R, Pimentel-Nunes P, Dinis-Ribeiro M, Libânio D. Gastrointestinal Microbiome - What We Need to Know in Clinical Practice. GE-PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2020; 27:336-351. [PMID: 32999906 DOI: 10.1159/000505036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/14/2019] [Indexed: 12/12/2022]
Abstract
Human gut microbiota plays an important role in individual health. When the balance between host and gut microbiota is disrupted, changes in microbiota composition and function occur, which is referred as dysbiosis. Environmental factors as diet, proton pump inhibitors, and antibiotics can lead to a permanent dysbiotic disruption. Clarification of these imbalances was made possible by recent advances in genome sequencing methods that supported acknowledgment of the interplay between microbiome and intestinal and extraintestinal disorders. This review focuses on the microbiota impact in inflammatory bowel disease, gastric cancer, colorectal cancer, nonalcoholic fatty liver disease (NAFLD), irritable bowel syndrome (IBS), and Clostridium difficile infection (CDI). Furthermore, novel therapies are summarized. Fecal microbiota transplant (FMT) is a successful and established therapy in recurrent CDI, and its application in other dysbiosis-related diseases is attracting enormous interest. Pre- and probiotics target microbial rebalance and have positive effects mainly in NAFLD, ulcerative colitis, IBS, and CDI patients. Promising anticarcinogenic effects have also been demonstrated in animal models. The literature increasingly describes microbial changes in many dysbiotic disorders and shows what needs to be treated. However, probiotics and FMT application in clinical practice suffers from a shortage of randomized controlled trials with standardized therapy regimens to support their recommendation.
Collapse
Affiliation(s)
- Raquel Ortigão
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Pedro Pimentel-Nunes
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal.,MEDCIDS - Department of Community Medicine, Information and Decision in Health, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal.,MEDCIDS - Department of Community Medicine, Information and Decision in Health, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diogo Libânio
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal.,MEDCIDS - Department of Community Medicine, Information and Decision in Health, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
68
|
Appaneal HJ, Caffrey AR, Beganovic M, Avramovic S, LaPlante KL. Predictors of Clostridioides difficile recurrence across a national cohort of veterans in outpatient, acute, and long-term care settings. Am J Health Syst Pharm 2019; 76:581-590. [PMID: 31361830 DOI: 10.1093/ajhp/zxz032] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE The greatest challenge in treating Clostridioides difficile infection (CDI) is disease recurrence, which occurs in about 20% of patients, usually within 30 days of treatment cessation. We sought to identify independent predictors of first recurrence among a national cohort of veterans with CDI. METHODS We conducted a case-control study among acute and long-term care Veterans Affairs (VA) inpatients and outpatients with a first CDI episode (positive stool sample for C. difficile toxin[s] and receipt of at least 2 days of CDI treatment) between 2010 and 2014. Cases experienced first recurrence within 30 days from the end of treatment. Controls were those without first recurrence matched 4:1 to cases on year, facility, and severity. Multivariable conditional logistic regression was used to identify predictors of first recurrence. RESULTS We identified 32 predictors of first recurrence among 974 cases and 3,896 matched controls. Significant predictors included medication use prior to (probiotics, fluoroquinolones, laxatives, third- or fourth-generation cephalosporins), during (first- or second-generation cephalosporins, penicillin/amoxicillin/ampicillin, third- and fourth-generation cephalosporins), and after CDI treatment (probiotics, any antibiotic, proton pump inhibitors [PPIs], and immunosuppressants). Other predictors included current biliary tract disease, malaise/fatigue, cellulitis/abscess, solid organ cancer, medical history of HIV, multiple myeloma, abdominal pain, and ulcerative colitis. CONCLUSION In a large national cohort of outpatient and acute and long-term care inpatients, treatment with certain antibiotics, PPIs, immunosuppressants, and underlying disease were among the most important risk factors for first CDI recurrence.
Collapse
Affiliation(s)
- Haley J Appaneal
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, and College of Pharmacy, University of Rhode Island, Kingston, RI
| | - Aisling R Caffrey
- Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, and College of Pharmacy, University of Rhode Island, Kingston, RI
| | - Maya Beganovic
- Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, and College of Pharmacy, University of Rhode Island, Kingston, RI
| | - Sanja Avramovic
- Health Administration and Policy, George Mason University, Fairfax, VA
| | - Kerry L LaPlante
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, and College of Pharmacy, University of Rhode Island, Kingston, RI
| |
Collapse
|
69
|
DAV131A Protects Hamsters from Lethal Clostridioides difficile Infection Induced by Fluoroquinolones. Antimicrob Agents Chemother 2019; 64:AAC.01196-19. [PMID: 31636067 DOI: 10.1128/aac.01196-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/08/2019] [Indexed: 02/04/2023] Open
Abstract
Fluoroquinolone treatments induce dysbiosis of the intestinal microbiota, resulting in loss of resistance to colonization by exogenous bacteria such as Clostridioides difficile that may cause severe diarrhea in humans and lethal infection in hamsters. We show here that DAV131A, a charcoal-based adsorbent, decreases the intestinal levels of the fluoroquinolone antibiotics levofloxacin and ciprofloxacin in hamsters, protects their intestinal microbiota, and prevents lethal infection by C. difficile.
Collapse
|
70
|
A Generalizable, Data-Driven Approach to Predict Daily Risk of Clostridium difficile Infection at Two Large Academic Health Centers. Infect Control Hosp Epidemiol 2019; 39:425-433. [PMID: 29576042 DOI: 10.1017/ice.2018.16] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE An estimated 293,300 healthcare-associated cases of Clostridium difficile infection (CDI) occur annually in the United States. To date, research has focused on developing risk prediction models for CDI that work well across institutions. However, this one-size-fits-all approach ignores important hospital-specific factors. We focus on a generalizable method for building facility-specific models. We demonstrate the applicability of the approach using electronic health records (EHR) from the University of Michigan Hospitals (UM) and the Massachusetts General Hospital (MGH). METHODS We utilized EHR data from 191,014 adult admissions to UM and 65,718 adult admissions to MGH. We extracted patient demographics, admission details, patient history, and daily hospitalization details, resulting in 4,836 features from patients at UM and 1,837 from patients at MGH. We used L2 regularized logistic regression to learn the models, and we measured the discriminative performance of the models on held-out data from each hospital. RESULTS Using the UM and MGH test data, the models achieved area under the receiver operating characteristic curve (AUROC) values of 0.82 (95% confidence interval [CI], 0.80-0.84) and 0.75 ( 95% CI, 0.73-0.78), respectively. Some predictive factors were shared between the 2 models, but many of the top predictive factors differed between facilities. CONCLUSION A data-driven approach to building models for estimating daily patient risk for CDI was used to build institution-specific models at 2 large hospitals with different patient populations and EHR systems. In contrast to traditional approaches that focus on developing models that apply across hospitals, our generalizable approach yields risk-stratification models tailored to an institution. These hospital-specific models allow for earlier and more accurate identification of high-risk patients and better targeting of infection prevention strategies. Infect Control Hosp Epidemiol 2018;39:425-433.
Collapse
|
71
|
Prolonged antimicrobial prophylaxis following cardiac device procedures increases preventable harm: insights from the VA CART program. Infect Control Hosp Epidemiol 2019; 39:1030-1036. [PMID: 30226128 DOI: 10.1017/ice.2018.170] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND The rate of cardiovascular implantable electronic device (CIED) infection is increasing coincident with an increase in the number of device procedures. Preprocedural antimicrobial prophylaxis reduces CIED infections; however, there is no evidence that prolonged postprocedural antimicrobials additionally reduce risk. Thus, we sought to quantify the harms associated with this approach. OBJECTIVE To measure the association between Clostridium difficile infection (CDI), acute kidney injury (AKI) and receipt of prolonged postprocedural antimicrobials. METHODS CIED procedures entered into the VA Clinical Assessment Reporting and Tracking Electrophysiology (CART-EP) database during fiscal years 2008-2016 were included. The primary outcome was 90-day incidence of CDI and the secondary outcome was the 7-day incidence of AKI. The primary exposure measure was duration of postprocedural antimicrobial therapy. Associations were measured using Cox-proportional hazards and binomial regression. RESULTS Prolonged postprocedural antimicrobial therapy was identified following 3,331 of 6,497 CIED procedures (51.3%), and the median duration of prophylaxis was 5 days. Prolonged postprocedural antimicrobial use was associated with increased risk of CDI (hazard ratio [HR], 2.90; 95% confidence interval [CI], 1.54-5.46). Of the 27 patients who developed CDI, 11 subsequently died. Postprocedural antimicrobial use with ≥2 antimicrobials was associated with an increased risk of AKI (OR, 4.16; 95% CI, 2.50-6.90). The impact was particularly significant when one of the dual agents prescribed was vancomycin (adjusted OR, 8.41; 95% CI, 5.53-12.79). CONCLUSIONS Prolonged antimicrobial prophylaxis following CIED procedures increases preventable harm; this practice should be discouraged in procedural settings such as the cardiac electrophysiology laboratory.
Collapse
|
72
|
Morrisette T, Van Matre AG, Miller MA, Mueller SW, Bajrovic V, Abidi MZ, Benamu E, Kaiser JN, Barber GR, Chase S, Tobin J, Fish DN, Gutman JA. Oral Vancomycin Prophylaxis as Secondary Prevention Against Clostridioides difficile Infection in the Hematopoietic Stem Cell Transplantation and Hematologic Malignancy Population. Biol Blood Marrow Transplant 2019; 25:2091-2097. [PMID: 31255741 DOI: 10.1016/j.bbmt.2019.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/27/2019] [Accepted: 06/24/2019] [Indexed: 01/02/2023]
Abstract
Clostridioides difficile infection (CDI) is a common complication in the hematopoietic stem cell transplantation (HSCT) and hematologic malignancy (HM) population. CDI is associated with increased hospital length of stay, health care and societal costs, morbidity, and mortality. Identifying strategies for secondary prevention of CDI is of extreme importance in the HSCT/HM population. In this study, our primary objective was to evaluate the effectiveness and safety of an oral vancomycin prophylaxis (OVP) protocol for secondary prevention of CDI in a retrospective cohort of adult autologous/allogeneic HSCT recipients and patients with HM who did not undergo HSCT with a first CDI episode treated with concomitant broad-spectrum antibiotics (BSA). Patients were diagnosed and treated for CDI as inpatients and/or outpatients and were divided into 2 groups based on a preprotocol versus postprotocol analysis: the OVP group, comprising patients who received planned monotherapy with oral vancomycin 125 mg every 6 hours for 14 days for a first episode of CDI and subsequently received OVP posttreatment and a no OVP (NOVP) group, comprising patients who received planned monotherapy with oral vancomycin 125 mg every 6 hours for 14 days for a first episode of CDI and subsequently did not receive OVP posttreatment. OVP was defined as vancomycin 125 mg every 12 hours for up to 7 days after BSA discontinuation. The primary endpoint was recurrent CDI (rCDI), defined as symptoms of loose stools/diarrhea with high clinical suspicion for CDI prompting empiric therapy within 60 days of completion of treatment/prophylaxis for the first CDI episode. The incidence of vancomycin-resistant enterococcal (VRE) infection and 60-day mortality were also compared between the 2 groups. Multivariate logistic regression was created from associated variables to identify independent associations with rCDI. A total of 50 patients were included, 21 in the OVP group (42%) and 29 in the NOVP group (58%). The mean patient age was 58 years, and the cohort was 60% male and 86% Caucasian. HSCT was performed in 60% of the patients, and 76% of CDI cases were diagnosed during hospitalization. The rate of rCDI was significantly lower in the OVP group compared with the NOVP group (5% [1 of 21] versus 35% [10 of 29]; P= .016), with no subsequent increase in VRE infection rate (14% [3 of 21] versus 10% [3 of 29]; P = .686). By multivariable logistic regression, rCDI was inversely associated with OVP (odds ratio [OR], .14; 95% confidence interval [CI], .007 to .994; P = .049) and directly associated with outpatient CDI diagnosis (OR, 8.72; 95% CI, 1.816 to 49.158; P = .007). No between-group differences were found in 60-day mortality (10% [2 of 21] for OVP versus 7% [2 of 29] for NOVP; P > 0.999). OVP appears to be safe and effective for secondary prevention of CDI in the HSCT/HM population. Prospective trials are needed to validate the effectiveness of OVP in this vulnerable population to prevent rCDI.
Collapse
Affiliation(s)
- Taylor Morrisette
- Department of Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado; Department of Pharmacy-Infectious Diseases, University of Colorado Hospital, Aurora, Colorado
| | - Amanda G Van Matre
- Department of Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado
| | - Matthew A Miller
- Department of Pharmacy-Infectious Diseases, University of Colorado Hospital, Aurora, Colorado
| | - Scott W Mueller
- Department of Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado
| | - Valida Bajrovic
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado
| | - Maheen Z Abidi
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado
| | - Esther Benamu
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado
| | - Jeffrey N Kaiser
- Department of Pharmacy-Blood Cancer and Bone Marrow Transplant, University of Colorado Hospital, Aurora, Colorado
| | - Gerard R Barber
- Department of Pharmacy-Infectious Diseases, University of Colorado Hospital, Aurora, Colorado
| | - Stephanie Chase
- Department of Pharmacy-Blood Cancer and Bone Marrow Transplant, University of Colorado Hospital, Aurora, Colorado
| | - Jennifer Tobin
- Department of Pharmacy-Blood Cancer and Bone Marrow Transplant, University of Colorado Hospital, Aurora, Colorado
| | - Douglas N Fish
- Department of Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, Colorado
| | - Jonathan A Gutman
- Department of Blood Cancer and Bone Marrow Transplant, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
73
|
Real-world effectiveness of infection prevention interventions for reducing procedure-related cardiac device infections: Insights from the veterans affairs clinical assessment reporting and tracking program. Infect Control Hosp Epidemiol 2019; 40:855-862. [PMID: 31159895 DOI: 10.1017/ice.2019.127] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To measure the association between receipt of specific infection prevention interventions and procedure-related cardiac implantable electronic device (CIED) infections. DESIGN Retrospective cohort with manually reviewed infection status. SETTING Setting: National, multicenter Veterans Health Administration (VA) cohort. PARTICIPANTS Sampling of procedures entered into the VA Clinical Assessment Reporting and Tracking-Electrophysiology (CART-EP) database from fiscal years 2008 through 2015. METHODS A sample of procedures entered into the CART-EP database underwent manual review for occurrence of CIED infection and other clinical/procedural variables. The primary outcome was 6-month incidence of CIED infection. Measures of association were calculated using multivariable generalized estimating equations logistic regression. RESULTS We identified 101 procedure-related CIED infections among 2,098 procedures (4.8% of reviewed sample). Factors associated with increased odds of infections included (1) wound complications (adjusted odds ratio [aOR], 8.74; 95% confidence interval [CI], 3.16-24.20), (2) revisions including generator changes (aOR, 2.4; 95% CI, 1.59-3.63), (3) an elevated international normalized ratio (INR) >1.5 (aOR, 1.56; 95% CI, 1.12-2.18), and (4) methicillin-resistant Staphylococcus colonization (aOR, 9.56; 95% CI, 1.55-27.77). Clinically effective prevention interventions included preprocedural skin cleaning with chlorhexidine versus other topical agents (aOR, 0.41; 95% CI, 0.22-0.76) and receipt of β-lactam antimicrobial prophylaxis versus vancomycin (aOR, 0.60; 95% CI, 0.37-0.96). The use of mesh pockets and continuation of antimicrobial prophylaxis after skin closure were not associated with reduced infection risk. CONCLUSIONS These findings regarding the real-world clinical effectiveness of different prevention strategies can be applied to the development of evidence-based protocols and infection prevention guidelines specific to the electrophysiology laboratory.
Collapse
|
74
|
Oral Beta-Lactamase Protects the Canine Gut Microbiome from Oral Amoxicillin-Mediated Damage. Microorganisms 2019; 7:microorganisms7050150. [PMID: 31137766 PMCID: PMC6560916 DOI: 10.3390/microorganisms7050150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/16/2019] [Accepted: 05/23/2019] [Indexed: 01/30/2023] Open
Abstract
Antibiotics damage the gut microbiome, which can result in overgrowth of pathogenic microorganisms and emergence of antibiotic resistance. Inactivation of antibiotics in the small intestine represents a novel strategy to protect the colonic microbiota. SYN-004 (ribaxamase) is a beta-lactamase formulated for oral delivery intended to degrade intravenously administered beta-lactam antibiotics in the gastrointestinal (GI) tract. The enteric coating of ribaxamase protects the enzyme from stomach acid and mediates pH-dependent release in the upper small intestine, the site of antibiotic biliary excretion. Clinical benefit was established in animal and human studies in which ribaxamase was shown to degrade ceftriaxone in the GI tract, thereby preserving the gut microbiome, significantly reducing Clostridioides difficile disease, and attenuating antibiotic resistance. To expand ribaxamase utility to oral beta-lactams, delayed release formulations of ribaxamase, SYN-007, were engineered to allow enzyme release in the lower small intestine, distal to the site of oral antibiotic absorption. Based on in vitro dissolution profiles, three SYN-007 formulations were selected for evaluation in a canine model of antibiotic-mediated gut dysbiosis. Dogs received amoxicillin (40 mg/kg, PO, TID) +/- SYN-007 (10 mg, PO, TID) for five days. Serum amoxicillin levels were measured after the first and last antibiotic doses and gut microbiomes were evaluated using whole genome shotgun sequence metagenomics analyses of fecal DNA prior to and after antibiotic treatment. Serum amoxicillin levels did not significantly differ +/- SYN-007 after the first dose for all SYN-007 formulations, while only one SYN-007 formulation did not significantly reduce systemic antibiotic concentrations after the last dose. Gut microbiomes of animals receiving amoxicillin alone displayed significant loss of diversity and emergence of antibiotic resistance genes. In contrast, for animals receiving amoxicillin + SYN-007, microbiome diversities were not altered significantly and the presence of antibiotic resistance genes was reduced. These data demonstrate that SYN-007 diminishes amoxicillin-mediated microbiome disruption and mitigates emergence and propagation of antibiotic resistance genes without interfering with antibiotic systemic absorption. Thus, SYN-007 has the potential to protect the gut microbiome by inactivation of beta-lactam antibiotics when administered by both oral and parenteral routes and to reduce emergence of antibiotic-resistant pathogens.
Collapse
|
75
|
Teng C, Reveles KR, Obodozie-Ofoegbu OO, Frei CR. Clostridium difficile Infection Risk with Important Antibiotic Classes: An Analysis of the FDA Adverse Event Reporting System. Int J Med Sci 2019; 16:630-635. [PMID: 31217729 PMCID: PMC6566741 DOI: 10.7150/ijms.30739] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/08/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction: Antibiotic use is an important risk factor for Clostridium difficile infection (CDI). Prior meta-analyses have identified antibiotics and antibiotic classes that pose the greatest risk for CDI; however, CDI epidemiology is constantly changing and contemporary analyses are needed. Objectives: The objective of this study was to evaluate the association between CDI and important antibiotic classes in recent years using the FDA Adverse Event Report System (FAERS). Methods: FAERS reports from January 1, 2015 to December 31, 2017 were analyzed. The Medical Dictionary for Regulatory Activities (MedDRA) was used to identify CDI cases. We computed the Reporting Odds Ratios (RORs) and corresponding 95% confidence intervals (95%CI) for the association between antibiotics and CDI. An association was considered statistically significant when the lower limit of the 95%CI was greater than 1. Results: A total of 2,042,801 reports (including 5,187 CDI reports) were considered, after inclusion criteria were applied. Lincosamides (e.g., clindamycin) had the greatest proportion of CDI reports, representing 10.4% of all lincosamide reports. CDI RORs (95%CI) for the antibiotic classes were (in descending order): lincosamides 46.95 (39.49-55.82), monobactams 29.97 (14.60-61.54), penicillin combinations 20.05 (17.39-23.12), carbapenems 19.16 (15.52-23.67), cephalosporins/ monobactams/carbapenems 17.28 (14.95-19.97), cephalosporins 15.33 (12.60-18.65), tetracyclines 7.54 (5.42-10.50), macrolides 5.80 (4.48-7.51), fluoroquinolones 4.94 (4.20-5.81), and trimethoprim-sulfonamides 3.32 (2.03-5.43). Conclusion: All antibiotic classes included in the study were significantly associated with CDI. Lincosamides (e.g., clindamycin) had the highest CDI ROR among the antibiotics evaluated in this study.
Collapse
Affiliation(s)
- Chengwen Teng
- Pharmacotherapy Division, College of Pharmacy, The University of Texas at Austin, San Antonio, TX, USA
- Pharmacotherapy Education and Research Center, Long School of Medicine, University of Texas Health-San Antonio, San Antonio, TX, USA
| | - Kelly R. Reveles
- Pharmacotherapy Division, College of Pharmacy, The University of Texas at Austin, San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Obiageri O. Obodozie-Ofoegbu
- Pharmacotherapy Division, College of Pharmacy, The University of Texas at Austin, San Antonio, TX, USA
- Pharmacotherapy Education and Research Center, Long School of Medicine, University of Texas Health-San Antonio, San Antonio, TX, USA
| | - Christopher R. Frei
- Pharmacotherapy Division, College of Pharmacy, The University of Texas at Austin, San Antonio, TX, USA
- University Health System, San Antonio, TX, USA
| |
Collapse
|
76
|
El-Matary W, Nugent Z, Yu BN, Lix LM, Targownik LE, Bernstein CN, Singh H. Trends and Predictors of Clostridium difficile Infection among Children: A Canadian Population-Based Study. J Pediatr 2019; 206:20-25. [PMID: 30448272 DOI: 10.1016/j.jpeds.2018.10.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/17/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To assess time trends in Clostridium difficile infection (CDI) rates, and predictors of CDIs, including recurrent CDIs, in children. STUDY DESIGN Data were extracted from Manitoba Health Provider Claims, and other population registry datasets from 2005 to 2015. CDI was identified from the Manitoba Health Public Health Branch Epidemiology and Surveillance population-based laboratory-confirmed CDI dataset. Children aged 2-17 years with CDI were matched by age, sex, area of residence, and duration of residence in Manitoba with children without CDI. The rates and time trends of CDIs using previously recommended definitions were determined. Predictors of CDI subtypes were determined using multivariable logistic regression models. Cox regression analysis was used to assess for the potential predictors of recurrent CDI. RESULTS Children with and without CDI were followed for 828 and 2753 persons-years, respectively. The overall CDI rate during the study period was 7.8 per 100 000 person-years. There was no significant change in CDI rates over the observation period. Comorbid conditions, more prevalent among children with CDI than matched controls, included Hirschsprung disease (P < .001) and inflammatory bowel disease (P < .0001). Recurrent CDIs (>2 occurrences) were responsible for 10% of CDI episodes (range, 2-6 infections). Predictors of recurrence included malignancy (hazard ratio, 3.0, 95% CI, 1.1-8.8), diabetes (hazard ratio, 4.8; 95% CI, 1.1-21.4), and neurodegenerative diseases (hazard ratio, 8.4; 95% CI, 1.9-37.5). CONCLUSIONS The incidence of CDI is stable among children in Manitoba. Children with Hirschsprung disease and inflammatory bowel disease are more susceptible to CDI, and those with malignancy, diabetes. and neurodegenerative disorders are more likely to develop recurrent CDI.
Collapse
Affiliation(s)
- Wael El-Matary
- Department of Pediatrics, University of Manitoba, Winnipeg, Manitoba, Canada; IBD Clinical and Research Center, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Zoann Nugent
- IBD Clinical and Research Center, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; CancerCare Manitoba, Department of Epidemiology and Cancer Registry, Winnipeg, Manitoba, Canada
| | - B Nancy Yu
- Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Public Health Branch, Manitoba Health, Seniors and Active Living, Winnipeg, Manitoba, Canada
| | - Lisa M Lix
- Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Laura E Targownik
- IBD Clinical and Research Center, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Charles N Bernstein
- IBD Clinical and Research Center, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Harminder Singh
- IBD Clinical and Research Center, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
77
|
Knight EM, Schiller DS, Fulman MK, Rastogi R. Long-Term Efficacy of Oral Vancomycin Prophylaxis for the Prevention of Clostridium difficile Recurrence. J Pharm Pract 2019; 33:633-639. [PMID: 30744479 DOI: 10.1177/0897190019825994] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Limited evidence suggests that prophylactic oral vancomycin may be beneficial in preventing Clostridium difficile infection (CDI) recurrence, but long-term efficacy is unknown. OBJECTIVE To evaluate the long-term efficacy of oral vancomycin prophylaxis (OVP) in preventing CDI recurrence in subjects who require subsequent antibiotic exposure. METHODS A retrospective cohort study was conducted at a community hospital. A total of 91 subjects with a history of CDI between January 2013 and December 2015 who had a subsequent hospitalization requiring systemic antibiotics within 12 months were evaluated. Thirty-two subjects who received prophylaxis with oral vancomycin were compared to 59 subjects who did not receive prophylaxis. RESULTS CDI recurrence within 12 months was significantly lower in subjects receiving OVP compared to those who did not receive OVP (6.3% vs 28.8%; odds ratio [OR]: 0.16; 95% confidence interval [CI]: 0.04-0.77; P = .011) including patients whose previous CDI was an initial episode (3.7% [1/27] vs 28.3% [15/53]; OR: 10.3; 95% CI: 1.28-82.6; P = .009). CONCLUSION Use of OVP in subjects with a history of CDI up to 12 months prior to subsequent antibiotic exposure appears to reduce the risk of CDI recurrence for up to 12 months.
Collapse
Affiliation(s)
- Emmanuel M Knight
- Department of Pharmacy Services, Montefiore Nyack Hospital, Nyack, NY, USA
| | - Daryl S Schiller
- Department of Pharmacy Services, Montefiore Nyack Hospital, Nyack, NY, USA
| | - Magda K Fulman
- Department of Pharmacy Services, Montefiore Nyack Hospital, Nyack, NY, USA
| | - Rupangi Rastogi
- Department of Pharmacy Services, Montefiore Nyack Hospital, Nyack, NY, USA
| |
Collapse
|
78
|
Connelly S, Fanelli B, Hasan NA, Colwell RR, Kaleko M. Oral Metallo-Beta-Lactamase Protects the Gut Microbiome From Carbapenem-Mediated Damage and Reduces Propagation of Antibiotic Resistance in Pigs. Front Microbiol 2019; 10:101. [PMID: 30804903 PMCID: PMC6370672 DOI: 10.3389/fmicb.2019.00101] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
Antibiotics can damage the gut microbiome, leading to serious adventitious infections and emergence of antibiotic resistant pathogens. Antibiotic inactivation in the GI tract represents a strategy to protect colonic microbiota integrity and reduce antibiotic resistance. Clinical utility of this approach was established when SYN-004 (ribaxamase), an orally-administered beta-lactamase, was demonstrated to degrade ceftriaxone in the GI tract and preserve the gut microbiome. Ribaxamase degrades penicillins and cephalosporin beta-lactams, but not carbapenems. To expand this prophylactic approach to include all classes of beta-lactam antibiotics, a novel carbapenemase, formulated for oral administration, SYN-006, was evaluated in a porcine model of antibiotic-mediated gut dysbiosis. Pigs (20 kg, n = 16) were treated with the carbapenem, ertapenem (ERT), (IV, 30 mg/kg, SID) for 4 days and a cohort (n = 8) also received SYN-006 (PO, 50 mg, QID), beginning the day before antibiotic administration. ERT serum levels were not statistically different in ERT and ERT + SYN-006 groups, indicating that SYN-006 did not alter systemic antibiotic levels. Microbiomes were evaluated using whole genome shotgun metagenomics analyses of fecal DNA collected prior to and after antibiotic treatment. ERT caused significant changes to the gut microbiome that were mitigated in the presence of SYN-006. In addition, SYN-006 attenuated emergence of antibiotic resistance, including encoded beta-lactamases and genes conferring resistance to a broad range of antibiotics such as aminoglycosides and macrolides. SYN-006 has the potential to become the first therapy designed to protect the gut microbiome from all classes of beta-lactam antibiotics and reduce emergence of carbapenem-resistant pathogens.
Collapse
Affiliation(s)
| | | | - Nur A Hasan
- CosmosID, Inc., Rockville, MD, United States
| | - Rita R Colwell
- CosmosID, Inc., Rockville, MD, United States.,University of Maryland Institute for Advanced Computer Studies, College Park, MD, United States
| | | |
Collapse
|
79
|
Klarin B, Larsson A, Molin G, Jeppsson B. Susceptibility to antibiotics in isolates of Lactobacillus plantarum RAPD-type Lp299v, harvested from antibiotic treated, critically ill patients after administration of probiotics. Microbiologyopen 2019; 8:e00642. [PMID: 29797784 PMCID: PMC6391266 DOI: 10.1002/mbo3.642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/11/2018] [Accepted: 03/23/2018] [Indexed: 01/21/2023] Open
Abstract
Recultured Lactobacillus plantarum 299v-like strains were tested regarding antibiotic susceptibility, and no decrease was detected. Antibiotics are frequently used to treat patients in intensive care units (ICUs) and are associated with a significant risk of selection of resistant bacterial strains. In particular, it is possible that genetic transfer of antibiotic resistance to the resident gastrointestinal flora, as well as to administered probiotics, may be increased in the ICU setting. The aim of the present investigation was to detect possible changes in antimicrobial susceptibility in reisolates of the probiotic strain Lactobacillus plantarum 299v (Lp299v) given to antibiotic treated, critically ill patients. Lp299v-like strains were identified in cultures of biopsies and fecal samples from 32 patients given the probiotic strain enterally in two previous ICU studies. The patients received a variety of antibiotics. Isolates with the same genomic RAPD profile (RAPD-type) as Lp299v were obtained to enable monitoring of antibiotic susceptibility by E-tests. Forty-two isolates, collected throughout the course of illness, were tested against 22 different antibiotics. No obvious decrease in susceptibility was found for 21 of the tested antibiotics. There was a tendency toward decreased susceptibility to ampicillin. The stable antibiotic susceptibility profiles of the Lp299v-like isolates studied here suggests this probiotic is less likely to acquire resistance when administered to critically ill patients treated with broad-spectrum antibiotics.
Collapse
Affiliation(s)
- Bengt Klarin
- Department of Anaesthesiology and Intensive CareLund University and Skåne University HospitalLundSweden
| | - Anders Larsson
- Department of Surgical SciencesSection of Anaesthesiology and Intensive CareUppsala University HospitalUppsalaSweden
| | - Göran Molin
- Applied Nutrition and Food ChemistryLund UniversityLundSweden
| | - Bengt Jeppsson
- Department of SurgeryLund University and Skåne University HospitalMalmöSweden
| |
Collapse
|
80
|
Abstract
The updated Infectious Disease Society of America guidelines for managing Clostridium difficile infections remove metronidazole as first-line therapy and add fecal microbiota transplants to the treatment options. This article reviews the new guidelines and strategies for diagnosis and infection control.
Collapse
|
81
|
Chia BY, Teo JQM, Lee W, Liew YX, Ee RPL, Chlebicki MP, Oon LLE, Kwa ALH. Do antimicrobial stewardship programme interventions reduce the rate of and protect against Clostridium difficile infection? J Glob Antimicrob Resist 2019; 17:312-315. [PMID: 30682564 DOI: 10.1016/j.jgar.2019.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Antimicrobial stewardship programmes (ASPs) have often been recommended as a viable solution to minimise the incidence of Clostridium difficile infection (CDI), which can be life-threatening. This study aimed to evaluate whether ASP interventions have contributed to reducing CDI rates. METHODS A retrospective review of ASP interventions issued from January 2013 to April 2014 was performed using data from the ASP database of Singapore General Hospital, a 1600-bed tertiary-care hospital in Singapore. A total of 283 interventions satisfied the inclusion criteria, of which commonly audited antibiotics were piperacillin/tazobactam (41.3%) and carbapenems (54.8%). Comparisons were made at 30days post-intervention between those with accepted or rejected interventions. The primary outcome was CDI incidence; secondary outcomes included length of hospitalisation post-intervention, 30-day mortality and CDI recurrence rate. RESULTS Whilst the median duration of antibiotic therapy was reduced by 2days (6days vs. 4 days; P<0.001), acceptance of ASP interventions did not alter primary CDI incidence at 30days (P=0.644) post-intervention. However, reduced CDI recurrence rates were observed for patients positive for CDI in the accepted patient group compared with the rejected group (0% vs. 37.5%; P=0.03), with no difference in CDI 30-day mortality between the two groups. CONCLUSION Intervention acceptance did not contribute to a significant reduction in CDI incidence but may be associated with lower recurrence rates, although further studies are required.
Collapse
Affiliation(s)
- Bih Yee Chia
- Department of Pharmacy, Singapore General Hospital, Singapore; Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | | | - Winnie Lee
- Department of Pharmacy, Singapore General Hospital, Singapore
| | - Yi Xin Liew
- Department of Pharmacy, Singapore General Hospital, Singapore
| | - Rachel Pui-Lai Ee
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | | | | | - Andrea Lay-Hoon Kwa
- Department of Pharmacy, Singapore General Hospital, Singapore; Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore; Emerging Infectious Disease Program, Duke-National University of Singapore Medical School, Singapore.
| |
Collapse
|
82
|
Identification, Characterization, and Formulation of a Novel Carbapenemase Intended to Prevent Antibiotic-Mediated Gut Dysbiosis. Microorganisms 2019; 7:microorganisms7010022. [PMID: 30654495 PMCID: PMC6352093 DOI: 10.3390/microorganisms7010022] [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] [Received: 12/03/2018] [Revised: 01/06/2019] [Accepted: 01/15/2019] [Indexed: 11/17/2022] Open
Abstract
Antibiotics can damage the gut microbiome leading to opportunistic infections and the emergence of antibiotic resistance. Microbiome protection via antibiotic inactivation in the gastrointestinal (GI) tract represents a strategy to limit antibiotic exposure of the colonic microbiota. Proof of concept for this approach was achieved with an orally-administered beta-lactamase enzyme, SYN-004 (ribaxamase), that was demonstrated to degrade ceftriaxone excreted into the GI tract and protect the gut microbiome from antibiotic-mediated dysbiosis. Ribaxamase efficiently degrades penicillin and cephalosporin beta-lactam antibiotics, but is not active against carbapenems. To expand this microbiome protection strategy to include all classes of beta-lactams, three distinct carbapenemases were evaluated for manufacturability, antibiotic degradation spectrum, and stability in human intestinal fluid. E. coli production strains were generated for P2A, a novel metallo-enzyme isolated from B. cereus, New Delhi metallo-beta-lactamase (NDM), and Klebsiella pneumoniae carbapenemase (KPC). While all three enzymes effectively inactivated a broad range of antibiotics, including penicillins, most cephalosporins, and carbapenems in vitro, only P2A retained biological activity when incubated with human chyme. As functional stability in the intestinal tract is a key requirement for an orally-delivered enzyme, P2A was chosen as a potential clinical candidate. An enteric formulation of P2A was developed, called SYN-006, that was inert under high acid conditions, with enzyme dissolution occurring at pH > 5.5. SYN-006 has the potential to expand microbiome protection via antibiotic inactivation to include all classes of beta-lactam antibiotics.
Collapse
|
83
|
Leedahl DD, Personett HA, Nagpal A, Barreto EF. Prevention of Clostridium difficile Infection in Critically Ill Adults. Pharmacotherapy 2019; 39:399-407. [PMID: 30506900 DOI: 10.1002/phar.2200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The incidence and severity of Clostridium difficile infection (CDI) remain high across intensive care units in the United States despite national efforts to decrease this escalating health care burden. Most published literature and guidelines address treatment rather than prevention, yet this approach may be too downstream to limit morbidity and mortality from the disease and its complications. Mechanisms to prevent CDI successfully include reducing modifiable risk factors and minimizing horizontal transmission of C. difficile spores between patients and the health care environment. Because CDI prevention is characterized by a bundled approach, it is difficult to quantify the individual impact of any one element; however, a number of patient- and facility-level strategies can be considered for CDI prevention. Robust hygiene strategies, diagnostic and antimicrobial stewardship, and particular prophylaxis maneuvers such as continuation of oral vancomycin or fidaxomicin in the setting of systemic antibiotics have all demonstrated benefit. The preventive roles of deprescribing acid suppressants, routine use of probiotics, or early fecal microbiota transplantation remain unclear. The focus of this review is to summarize the evidence related to primary and secondary CDI prevention in critically ill adults and provide a concise implementation pathway for clinicians and policymakers.
Collapse
Affiliation(s)
- David D Leedahl
- Pharmacy Services, Sanford Medical Center, Fargo, North Dakota
| | | | - Avish Nagpal
- Infectious Diseases, Sanford Medical Center, Fargo, North Dakota
| | - Erin F Barreto
- Department of Pharmacy, Mayo Clinic, Rochester, Minnesota.,Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
84
|
Lopes Cançado GG, Silveira Silva RO, Rupnik M, Nader AP, Starling de Carvalho J, Miana de Mattos Paixão G, Martins Resende BA, Faria Lobato FC, Vilela EG. Clinical epidemiology of Clostridium difficile infection among hospitalized patients with antibiotic-associated diarrhea in a university hospital of Brazil. Anaerobe 2018; 54:65-71. [DOI: 10.1016/j.anaerobe.2018.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/02/2018] [Accepted: 08/12/2018] [Indexed: 02/08/2023]
|
85
|
Silvetti S, Landoni G. Is Clostridium difficile the new bugaboo after cardiac surgery? J Thorac Dis 2018; 10:S3278-S3280. [PMID: 30370137 DOI: 10.21037/jtd.2018.08.99] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Simona Silvetti
- Department of Pediatric Intensive Care Unit and Cardiac Anesthesia, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Giovanni Landoni
- Department of Cardiac Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
86
|
Antibiotic treatment of infections caused by carbapenem-resistant Gram-negative bacilli: an international ESCMID cross-sectional survey among infectious diseases specialists practicing in large hospitals. Clin Microbiol Infect 2018; 24:1070-1076. [DOI: 10.1016/j.cmi.2018.01.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 01/02/2023]
|
87
|
Huston JM, Zuckerbraun BS, Moore LJ, Sanders JM, Duane TM. Antibiotics versus No Antibiotics for the Treatment of Acute Uncomplicated Diverticulitis: Review of the Evidence and Future Directions. Surg Infect (Larchmt) 2018; 19:648-654. [DOI: 10.1089/sur.2018.115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Jared M. Huston
- Departments of Surgery and Science Education, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Brian S. Zuckerbraun
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Laura J. Moore
- Department of Surgery, The University of Texas McGovern Medical School, Houston, Texas
| | - James M. Sanders
- Department of Pharmacy and John Peter Smith Health Network, Fort Worth, Texas
| | - Therese M. Duane
- Department of Surgery, John Peter Smith Health Network, Fort Worth, Texas
| |
Collapse
|
88
|
Alexander S, Fisher BT, Gaur AH, Dvorak CC, Villa Luna D, Dang H, Chen L, Green M, Nieder ML, Fisher B, Bailey LC, Wiernikowski J, Sung L. Effect of Levofloxacin Prophylaxis on Bacteremia in Children With Acute Leukemia or Undergoing Hematopoietic Stem Cell Transplantation: A Randomized Clinical Trial. JAMA 2018; 320:995-1004. [PMID: 30208456 PMCID: PMC6143098 DOI: 10.1001/jama.2018.12512] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 08/03/2018] [Indexed: 11/14/2022]
Abstract
Importance Bacteremia causes considerable morbidity among children with acute leukemia and those undergoing hematopoietic stem cell transplantation (HSCT). There are limited data on the effect of antibiotic prophylaxis in children. Objective To determine the efficacy and risks of levofloxacin prophylaxis in children receiving intensive chemotherapy for acute leukemia or undergoing HSCT. Design, Setting, and Participants In this multicenter, open-label, randomized trial, patients (6 months-21 years) receiving intensive chemotherapy were enrolled (September 2011-April 2016) in 2 separate groups-acute leukemia, consisting of acute myeloid leukemia or relapsed acute lymphoblastic leukemia, and HSCT recipients-at 76 centers in the United States and Canada, with follow-up completed September 2017. Interventions Patients with acute leukemia were randomized to receive levofloxacin prophylaxis for 2 consecutive cycles of chemotherapy (n = 100) or no prophylaxis (n = 100). Those undergoing HSCT were randomized to receive levofloxacin prophylaxis during 1 HSCT procedure (n = 210) or no prophylaxis (n = 214). Main Outcomes and Measures The primary outcome was the occurrence of bacteremia during 2 chemotherapy cycles (acute leukemia) or 1 transplant procedure (HSCT). Secondary outcomes included fever and neutropenia, severe infection, invasive fungal disease, Clostridium difficile-associated diarrhea, and musculoskeletal toxic effects. Results A total of 624 patients, 200 with acute leukemia (median [interquartile range {IQR}] age, 11 years [6-15 years]; 46% female) and 424 undergoing HSCT (median [IQR] age, 7 years [3-14]; 38% female), were enrolled. Among 195 patients with acute leukemia, the likelihood of bacteremia was significantly lower in the levofloxacin prophylaxis group than in the control group (21.9% vs 43.4%; risk difference, 21.6%; 95% CI, 8.8%-34.4%, P = .001), whereas among 418 patients undergoing HSCT, the risk of bacteremia was not significantly lower in the levofloxacin prophylaxis group (11.0% vs 17.3%; risk difference, 6.3%; 95% CI, 0.3%-13.0%; P = .06). Fever and neutropenia were less common in the levofloxacin group (71.2% vs 82.1%; risk difference, 10.8%; 95% CI, 4.2%-17.5%; P = .002). There were no significant differences in severe infection (3.6% vs 5.9%; risk difference, 2.3%; 95% CI, -1.1% to 5.6%; P = .20), invasive fungal disease (2.9% vs 2.0%; risk difference, -1.0%; 95% CI, -3.4% to 1.5%, P = .41), C difficile-associated diarrhea (2.3% vs 5.2%; risk difference, 2.9%; 95% CI, -0.1% to 5.9%; P = .07), or musculoskeletal toxic effects at 2 months (11.4% vs 16.3%; risk difference, 4.8%; 95% CI, -1.6% to 11.2%; P = .15) or at 12 months (10.1% vs 14.4%; risk difference, 4.3%; 95% CI, -3.4% to 12.0%; P = .28) between the levofloxacin and control groups. Conclusions and Relevance Among children with acute leukemia receiving intensive chemotherapy, receipt of levofloxacin prophylaxis compared with no prophylaxis resulted in a significant reduction in bacteremia. However, there was no significant reduction in bacteremia for levofloxacin prophylaxis among children undergoing HSCT.
Collapse
Affiliation(s)
| | - Brian T. Fisher
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Aditya H. Gaur
- St Jude Children's Research Hospital, Memphis, Tennessee
| | | | | | - Ha Dang
- University of Southern California, Los Angeles, California
| | - Lu Chen
- City of Hope, Duarte, California
| | - Michael Green
- Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburg, Pennsylvania
| | | | - Beth Fisher
- Children's Healthcare of Atlanta, Egleston, Atlanta, Georgia
| | | | | | - Lillian Sung
- The Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluation Sciences, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
89
|
Comparison of qPCR versus culture for the detection and quantification of Clostridium difficile environmental contamination. PLoS One 2018; 13:e0201569. [PMID: 30161136 PMCID: PMC6116935 DOI: 10.1371/journal.pone.0201569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Contaminated surfaces serve as an important reservoir for Clostridium difficile transmission. Current strategies to detect environmental contamination of C. difficile rely heavily on culture, and often only indicate presence versus absence of spores. The goal of this study was to compare quantitative PCR (qPCR) to culture for the detection and quantification of C. difficile from inert surfaces. First, we compared the limit of detection (LOD) of a 16S rRNA gene and toxin B gene qPCR assay for detection of C. difficile in solution. Second, we compared the LODs of 16S rRNA gene qPCR versus culture for detection of C. difficile from surfaces. Solution experiments were performed by direct seeding of spores into neutralizing broth, whereas surface experiments involved seeding of spores onto plastic test surfaces, and recovery using sponge swabs. Both experiments were conducted using spores expressing short (NAP1) and long (NAP4) hair lengths. Combining data from both strains, the overall LOD for C. difficile cells in solution was 1.4 cells for 16S rRNA gene and 23.6 cells for toxin B gene qPCR (p<0.001). The overall LOD for C. difficile cells from surfaces was 17.1 cells for 16S rRNA gene qPCR and 54.5 cells for culture (p = 0.05), and was not statistically different between strains for each method (p = 0.52). Overall, the proportion of C. difficile cells recovered from surfaces was good when detected by 16S rRNA gene qPCR and culture (qPCR: 76%, culture: 67%, p = 0.36), but, 16S rRNA gene qPCR was capable of detecting lower levels of surface contamination. Future work attempting to measure the presence of C. difficile on environmental surfaces should consider using qPCR.
Collapse
|
90
|
Increased environmental sample area and recovery ofClostridium difficilespores from hospital surfaces by quantitative PCR and enrichment culture. Infect Control Hosp Epidemiol 2018; 39:917-923. [DOI: 10.1017/ice.2018.103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractObjectiveClostridium difficilespores play an important role in transmission and can survive in the environment for several months. Optimal methods for measuring environmentalC. difficileare unknown. We sought to determine whether increased sample surface area improved detection ofC. difficilefrom environmental samples.SettingSamples were collected from 12 patient rooms in a tertiary-care hospital in Toronto, Canada.MethodsSamples represented small surface-area and large surface-area floor and bedrail pairs from single-bed rooms of patients with low (without prior antibiotics), medium (with prior antibiotics), and high (C. difficileinfected) shedding risk. Presence ofC. difficilein samples was measured using quantitative polymerase chain reaction (qPCR) with targets on the 16S rRNA and toxin B genes and using enrichment culture.ResultsOf the 48 samples, 64·6% were positive by 16S qPCR (geometric mean, 13·8 spores); 39·6% were positive by toxin B qPCR (geometric mean, 1·9 spores); and 43·8% were positive by enrichment culture. By 16S qPCR, each 10-fold increase in sample surface area yielded 6·6 times (95% CI, 3·2–13) more spores. Floor surfaces yielded 27 times (95% CI, 4·9–181) more spores than bedrails, and rooms ofC. difficile–positive patients yielded 11 times (95% CI, 0·55–164) more spores than those of patients without prior antibiotics. Toxin B qPCR and enrichment culture returned analogous findings.ConclusionsClostridium difficilespores were identified in most floor and bedrail samples, and increased surface area improved detection. Future research aiming to understand the role of environmentalC. difficilein transmission should prefer samples with large surface areas.
Collapse
|
91
|
Abstract
Each year in the United States, billions of dollars are spent combating almost half a million Clostridium difficile infections (CDIs) and trying to reduce the ∼29,000 patient deaths in which C. difficile has an attributed role. In Europe, disease prevalence varies by country and level of surveillance, though yearly costs are estimated at €3 billion. One factor contributing to the significant health care burden of C. difficile is the relatively high frequency of recurrent CDIs. Recurrent CDI, i.e., a second episode of symptomatic CDI occurring within 8 weeks of successful initial CDI treatment, occurs in ∼25% of patients, with 35 to 65% of these patients experiencing multiple episodes of recurrent disease. Using microbial communities to treat recurrent CDI, either as whole fecal transplants or as defined consortia of bacterial isolates, has shown great success (in the case of fecal transplants) or potential promise (in the case of defined consortia of isolates). This review will briefly summarize the epidemiology and physiology of C. difficile infection, describe our current understanding of how fecal microbiota transplants treat recurrent CDI, and outline potential ways that knowledge can be used to rationally design and test alternative microbe-based therapeutics.
Collapse
|
92
|
Vardakas KZ, Kalimeris GD, Triarides NA, Falagas ME. An update on adverse drug reactions related to β-lactam antibiotics. Expert Opin Drug Saf 2018; 17:499-508. [DOI: 10.1080/14740338.2018.1462334] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Konstantinos Z. Vardakas
- Alfa Institute of Biomedical Sciences, Athens, Greece
- Department of Medicine, Henry Dunant Hospital Center, Athens, Greece
| | - Georgios D. Kalimeris
- Alfa Institute of Biomedical Sciences, Athens, Greece
- Department of Medicine, Henry Dunant Hospital Center, Athens, Greece
| | - Nikolaos A. Triarides
- Alfa Institute of Biomedical Sciences, Athens, Greece
- Department of Medicine, Henry Dunant Hospital Center, Athens, Greece
| | - Matthew E. Falagas
- Alfa Institute of Biomedical Sciences, Athens, Greece
- Department of Medicine, Henry Dunant Hospital Center, Athens, Greece
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
93
|
Bartoletti M, Tedeschi S, Pascale R, Raumer L, Maraolo AE, Palmiero G, Tumietto F, Cristini F, Ambretti S, Giannella M, Lewis RE, Viale P. Differences in the rate of carbapenem-resistant Enterobacteriaceae colonisation or Clostridium difficile infection following frontline treatment with tigecycline vs. meropenem for intra-abdominal infections. Int J Antimicrob Agents 2018; 51:516-521. [PMID: 29410250 DOI: 10.1016/j.ijantimicag.2018.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/29/2017] [Accepted: 01/13/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES We hypothesised that treatment with a tigecycline-based antimicrobial regimen for intra-abdominal infection (IAI) could be associated with lower rates of subsequent carbapenem-resistant Enterobacteriaceae (CRE) colonisation or Clostridium difficile infection (CDI) compared with a meropenem-based regimen. METHODS We performed a retrospective, single-centre, matched (1:1) cohort analysis of all patients who received at least 5 days of empirical or targeted tigecycline (TIG)- or meropenem (MER)-based treatment regimens for IAI over a 50-month period. Patients with previous CRE colonisation and CDI were excluded. Risk factors for CRE and CDI were assessed with a Cox regression model that included treatment duration as a time-dependent variable. Thirty-day mortality was assessed with Kaplan-Meier curves. RESULTS We identified 168 TIG-treated and 168 MER-treated patients. The cumulative incidence rate ratio of CDI was 10-fold lower in TIG-treated vs. MER-treated patients (incidence rate ratio [IRR] 0.10/1000 patient-days, 95%CI 0.002-0.72, P = 0.007), but similar incidence rates were found for CRE colonisation (IRR 1.39/1000 patient-days, 95%CI 0.68-2.78, P = 0.36). In a multivariate Cox regression model, the receipt of a TIG- vs. MER-based regimen was associated with significantly lower rates of CDI (HR 0.07, 95%CI 0.03-0.71, P = 0.02), but not CRE (HR 1.12, 95% CI 0.45-2.83, P = 0.80). All-cause 30-day mortality was similar in the two groups (P = 0.46). CONCLUSION TIG-based regimens for IAI were associated with a 10-fold lower incidence of CDI compared with MER-based regimens, but there was no difference in the incidence of CRE colonisation.
Collapse
Affiliation(s)
- Michele Bartoletti
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Sara Tedeschi
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Renato Pascale
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Luigi Raumer
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Alberto Enrico Maraolo
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, via Sergio Pansini 5, 80131, Naples, Italy
| | - Giulia Palmiero
- Section of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples Federico II, via Sergio Pansini 5, 80131, Naples, Italy
| | - Fabio Tumietto
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Francesco Cristini
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Simone Ambretti
- Operative Unit of Clinical Microbiology, S. Orsola-Malpighi Hospital, Via Massarenti 9, 40138, Bologna, Italy
| | - Maddalena Giannella
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Russell Edward Lewis
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Pierluigi Viale
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| |
Collapse
|
94
|
Duarte-Chavez R, Wojda TR, Zanders TB, Geme B, Fioravanti G, Stawicki SP. Early Results of Fecal Microbial Transplantation Protocol Implementation at a Community-based University Hospital. J Glob Infect Dis 2018; 10:47-57. [PMID: 29910564 PMCID: PMC5987372 DOI: 10.4103/jgid.jgid_145_17] [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] [Indexed: 12/14/2022] Open
Abstract
Introduction: Clostridium difficile (CD) is a serious and increasingly prevalent healthcare-associated infection. The pathogenesis of CD infection (CDI) involves the acquisition of CD with a concurrent disruption of the native gut flora. Antibiotics are a major risk although other contributing factors have also been identified. Clinical management combines discontinuation of the offending antibiotic, initiation of CD-specific antibiotic therapy, probiotic agent use, fecal microbiota transplantation (FMT), and surgery as the “last resort” option. The aim of this study is to review short-term clinical results following the implementation of FMT protocol (FMTP) at our community-based university hospital. Methods: After obtaining Institutional Review Board and Infection Control Committee approvals, we implemented an institution-wide FMTP for patients diagnosed with CDI. Prospective tracking of all patients receiving FMT between July 1, 2015, and February 1, 2017, was conducted using REDCap™ electronic data capture system. According to the FMTP, indications for FMT included (a) three or more CDI recurrences, (b) two or more hospital admissions with severe CDI, or (c) first episode of complicated CDI (CCDI). Risk factors for initial infection and for treatment failure were assessed. Patients were followed for at least 3 months to monitor for cure/failure, relapse, and side effects. Frozen 250 mL FMT samples were acquired from OpenBiome (Somerville, MA, USA). After 4 h of thawing, the liquid suspension was applied using colonoscopy, beginning with terminal ileum and proceeding distally toward mid-transverse colon. Monitored clinical parameters included disease severity (Hines VA CDI Severity Score or HVCSS), concomitant medications, number of FMT treatments, non-FMT therapies, cure rates, and mortality. Descriptive statistics were utilized to outline the study results. Results: A total of 35 patients (mean age 58.5 years, 69% female) were analyzed, with FMT-attributable primary cure achieved in 30/35 (86%) cases. Within this subgroup, 2/30 (6.7%) patients recurred and were subsequently cured with long-term oral vancomycin. Among five primary FMT failures (14% total sample), 3 (60%) achieved medical cure with long-term oral vancomycin therapy and 2 (40%) required colectomy. For the seven patients who either failed FMT or recurred, long-term vancomycin therapy was curative in all but two cases. For patients with severe CDI (HVCSS ≥3), primary and overall cure rates were 6/10 (60%) and 8/10 (80%), respectively. Patients with CCDI (n = 4) had higher HVCSS (4 vs. 3) and a mortality of 25%. Characteristics of patients who failed initial FMT included older age (70 vs. 57 years), female sex (80% vs. 67%), severe CDI (80% vs. 13%), and active opioid use during the initial infection (60% vs. 37%) and at the time of FMT (60% vs. 27%). The most commonly reported side effect of FMT was loose stools. Conclusions: This pilot study supports the efficacy and safety of FMT administration for CDI in the setting of a community-based university hospital. Following FMTP implementation, primary (86%) and overall (94%) nonsurgical cure rates were similar to those reported in other studies. The potential role of opioids as a modulator of CDI warrants further clinical investigation.
Collapse
Affiliation(s)
- Rodrigo Duarte-Chavez
- Department of Internal Medicine, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Thomas R Wojda
- Department of Family Medicine, Warren Hospital, St. Luke's University Health Network, Phillipsburg, NJ, USA
| | - Thomas B Zanders
- Division of Pulmonary/Critical Care Medicine, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Berhanu Geme
- Division of Gastroenterology, St. Luke's University Health Network, Bethlehem, PA, USA
| | - Gloria Fioravanti
- Department of Internal Medicine, St. Luke's University Health Network, Bethlehem, PA, USA
| | | |
Collapse
|
95
|
Zarandi ER, Mansouri S, Nakhaee N, Sarafzadeh F, Moradi M. Effect of sub-MIC of vancomycin and clindamycin alone and in combination with ceftazidime on Clostridium difficile surface layer protein A (slpA) gene expression. Microb Pathog 2017; 111:163-167. [PMID: 28867619 DOI: 10.1016/j.micpath.2017.08.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 08/30/2017] [Accepted: 08/30/2017] [Indexed: 11/15/2022]
Abstract
Clostridium difficile (C.difficile) infection is often established in the presence of antibiotics and probably antibiotics can influence surface layer protein A (slpA) expression as a colonization factor. The aim of this study is to evaluate the effect of vancomycin (VAN), clindamycin (CLI) alone and in combination with ceftazidime (CAZ) on slpA gene expression to determine whether such antibiotics can have any effect on slpA expression. About ∼106 CFU/mL was inoculated to medium containing an appropriate concentration of antibiotics alone and in combination. After 24 and 48 h incubation under anaerobic condition, 3 mL of culture was excluded and centrifuged in 8000 × g per 3 min. The pellet was washed and used for RNA extraction. The RNA extraction, Dnase I treatment and cDNA synthesis was performed by RNA extraction, Dnase I, and cDNA synthesis kits, respectively. The real-time PCR were carried out by sybrGreen methods and data were analyzed based on comparative ΔΔCT. All antibiotics alone and in combination, except VAN/CAZ in clinical isolate, decreased the level of slpA gene expression in the 24-h incubation. While the expression profile of slpA was different in the 48-h incubation period. The VAN and CLI decreased the slpA expression, although the template of expression is closed to control medium. CAZ alone and in combination increased slpA expression. C. difficile may down-regulate slpA expression in the early stages of growth in sub-inhibitory concentration of antibiotics. But, over time C. difficile increases or over expresses the slpA expression level. Consequently C. difficile binds strongly to epithelial cells and continues to survive in the presence of sub-MIC concentration of antibiotics. This effect is observed especially with regard to CAZ and probably other third generation cephalosporins or in combination therapy with VAN or CLI, which are prescribed in the clinic. CAZ can interfere with the anti-down regulatory feature of VAN, CLI, and maybe other antibiotics.
Collapse
Affiliation(s)
- Ebrahim Rezazadeh Zarandi
- Department of Microbiology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahla Mansouri
- Department of Microbiology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nouzar Nakhaee
- Department of Community Medicine, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Farhad Sarafzadeh
- Department of Infectious Diseases, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Moradi
- Department of Microbiology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
96
|
Affiliation(s)
- Hemant Goyal
- Mercer University School of Medicine, Macon, Georgia, USA
| | - Harold Katner
- Mercer University School of Medicine, Macon, Georgia, USA
| |
Collapse
|
97
|
Zarandi ER, Mansouri S, Nakhaee N, Sarafzadeh F, Moradi M. Toxin production of Clostridium difficile in sub-MIC of vancomycin and clindamycin alone and in combination with ceftazidime. Microb Pathog 2017; 107:249-253. [PMID: 28286152 DOI: 10.1016/j.micpath.2017.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/26/2017] [Accepted: 03/06/2017] [Indexed: 02/01/2023]
Abstract
Toxin production in Clostridium difficile (C. difficile) is a key process for induction of diarrhea. Several factors such as sub-MIC of antibiotics impact on toxin production. The aim of this research is investigation of sub-minimum inhibitory concentration (sub-MIC) of vancomycin (VAN), clindamycin (CLI) separately and in combination with ceftazidime (CAZ) on toxin production in C. difficile. About ∼106 colony forming units (CFU) from 18-h culture of C. difficile ATCC 9689 and clinical isolates A+/B+/CTD-, were cultured anaerobically in the pre-reduced medium with appropriate concentration of separated and in combination antibiotics. After 24 and 48 h, 1 mL of culture was removed, centrifuged and the supernatant stored at-70 °C for later use. The evaluation of toxin production was carried out by the ELISA technique. All antibiotics alone and in combination formats inhibited toxin production over a period of 24 h. This effect is also observed in presence of VAN and CLI during a period of 48 h. Over a 4 h period, CAZ increased toxin production alone and in combination, especially with CLI. The data showed VAN and CLI decrease the level of toxins. In contrast, the CAZ not only increases the level of produced toxin, but also can interfere with VAN and CLI. Based on the results, combination therapy which is performed for treatment or prevention of other infections may cause toxin production and probably the severity of C. difficile AAD to increase.
Collapse
Affiliation(s)
- Ebrahim Rezazadeh Zarandi
- Department of Microbiology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahla Mansouri
- Department of Microbiology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nouzar Nakhaee
- Department of Community Medicine, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Farhad Sarafzadeh
- Department of Infectious Diseases, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Moradi
- Department of Microbiology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
98
|
Reply to "Antibiotics, intestinal dysbiosis and risk of celiac disease" by Hakim Rahmoune et al. [Digestive and Liver Disease]. Dig Liver Dis 2017; 49:106-107. [PMID: 27840057 DOI: 10.1016/j.dld.2016.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 10/14/2016] [Indexed: 12/11/2022]
|
99
|
Kullin B, Wojno J, Abratt V, Reid SJ. Toxin A-negative toxin B-positive ribotype 017 Clostridium difficile is the dominant strain type in patients with diarrhoea attending tuberculosis hospitals in Cape Town, South Africa. Eur J Clin Microbiol Infect Dis 2016; 36:163-175. [DOI: 10.1007/s10096-016-2790-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022]
|