Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Antimicrob Agents Chemother ; 68(3): e0162123, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38364016

ABSTRACT

Antimicrobial resistance is emerging in clinical strains of Clostridioides difficile. Ibezapolstat (IBZ) is a DNA polymerase IIIC inhibitor that has completed phase II clinical trials. IBZ has potent in vitro activity against wild-type, susceptible strains but its effect on C. difficile strains with reduced susceptibility to metronidazole (MTZ), vancomycin (VAN), or fidaxomicin (FDX) has not been tested. The primary objective of this study was to test the antibacterial properties of IBZ against multidrug-resistant C. difficile strains. The in vitro activity, bactericidal, and time-kill activity of IBZ versus comparators were evaluated against 100 clinical strains of which 59 had reduced susceptibility to other C. difficile antibiotics. Morphologic changes against a multidrug resistance strain were visualized by light and scanning electron microscopy. The overall IBZ MIC50/90 values (µg/mL) for evaluated C. difficile strains were 4/8, compared with 2/4 for VAN, 0.5/1 for FDX, and 0.25/4 for MTZ. IBZ MIC50/90 values did not differ based on non-susceptibility to antibiotic class or number of classes to which strains were non-susceptible. IBZ bactericidal activity was similar to the minimum inhibitory concentration (MIC) and maintained in wild-type and non-susceptible strains. Time-kill assays against two laboratory wild-type and two clinical non-susceptible strains demonstrated sustained IBZ activity despite reduced killing by comparator antibiotics for IBZ and VAN non-susceptible strains. Microscopy visualized increased cell lengthening and cellular damage in multidrug-resistant strains exposed to IBZ sub-MIC concentrations. This study demonstrated the potent antibacterial activity of IBZ against a large collection of C. difficile strains including multidrug-resistant strains. This study highlights the therapeutic potential of IBZ against multidrug-resistant strains of C. difficile.


Subject(s)
Anti-Infective Agents , Clostridioides difficile , Clostridium Infections , Purine Nucleosides , Humans , Clostridioides , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents/pharmacology , Vancomycin/pharmacology , Vancomycin/therapeutic use , Metronidazole/pharmacology , Metronidazole/therapeutic use , Fidaxomicin/pharmacology , Fidaxomicin/therapeutic use , Microbial Sensitivity Tests
2.
Antimicrob Agents Chemother ; 67(5): e0156322, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37093023

ABSTRACT

Clostridioides difficile infection (CDI) causes substantial morbidity and mortality worldwide with limited antibiotic treatment options. Ridinilazole is a precision bisbenzimidazole antibiotic being developed to treat CDI and reduce unacceptably high rates of infection recurrence in patients. Although in late clinical development, the precise mechanism of action by which ridinilazole elicits its bactericidal activity has remained elusive. Here, we present conclusive biochemical and structural data to demonstrate that ridinilazole has a primary DNA binding mechanism, with a co-complex structure confirming binding to the DNA minor groove. Additional RNA-seq data indicated early pleiotropic changes to transcription, with broad effects on multiple C. difficile compartments and significant effects on energy generation pathways particularly. DNA binding and genomic localization was confirmed through confocal microscopy utilizing the intrinsic fluorescence of ridinilazole upon DNA binding. As such, ridinilazole has the potential to be the first antibiotic approved with a DNA minor groove binding mechanism of action.


Subject(s)
Clostridioides difficile , Clostridium Infections , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/genetics , Pyridines/pharmacology , Clostridium Infections/drug therapy
3.
Anaerobe ; 79: 102682, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36580991

ABSTRACT

Three strictly anaerobic strains of Escherichia coli were misidentified as Fusobacterium mortiferum, due to a deletion of the hemB gene which is involved in anaerobic respiration. An unusual antimicrobial susceptibility pattern sparked the further diagnostic strategies that eventually identified these strains as true anaerobic E. coli This phenomenon is more common than appreciated and can have an impact on clinical practice including persistent and relapsing infections.


Subject(s)
Fusobacteria , Fusobacterium Infections , Humans , Anaerobiosis , Escherichia coli/genetics , Fusobacterium Infections/microbiology
4.
Antimicrob Agents Chemother ; 66(8): e0224421, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35862742

ABSTRACT

Reduction of Clostridioides difficile infection (CDI) recurrence is an essential endpoint for CDI-directed antibiotic development that is often not evaluated until Phase III trials. The purpose of this project was to use a functional and metagenomic approach to predict the potential anti-CDI recurrence effect of ibezapolstat, a DNA polymerase IIIC inhibitor, in clinical development for CDI. As part of the Phase I ibezapolstat clinical study, stool samples were collected from 22 healthy volunteers, who were given either ibezapolstat or vancomycin. Stool samples were evaluated for microbiome changes and bile acid concentrations. Ibezapolstat 450 mg and vancomycin, but not ibezapolstat 300 mg, showed statistically significant changes in alpha diversity over time compared to that of a placebo. Beta diversity changes confirmed that microbiota were significantly different between study groups. Vancomycin had a more wide-ranging effect on the microbiome, characterized by an increased proportion of Gammaproteobacteria. Ibezapolstat demonstrated an increased proportion of Actinobacteria, including the Bifidobacteriaceae family. Using a linear regression analysis, vancomycin was associated with significant increases in primary bile acids as well as primary:secondary bile acid ratios. An overabundance of Enterobacteriaceae was most highly correlated with primary bile acid concentrations (r = 0.63; P < 0.0001). Using Phase I healthy volunteer samples, beneficial changes suggestive of a lower risk of CDI recurrence were associated with ibezapolstat compared to vancomycin. This novel omics approach may allow for better and earlier prediction of anti-CDI recurrence effects for antibiotics in the clinical development pipeline.


Subject(s)
Actinobacteria , Clostridioides difficile , Clostridium Infections , Actinobacteria/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bile Acids and Salts/therapeutic use , Clostridioides difficile/genetics , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Humans , Vancomycin/pharmacology , Vancomycin/therapeutic use
5.
Anaerobe ; 72: 102440, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34461273

ABSTRACT

BACKGROUND: Although Clostridioides difficile surveillance often identifies emerging strains, clinical outcome evaluations are rarely performed. Ribotype (RT) 106 is a commonly isolated C. difficile strain worldwide; however, studies investigating RT 106 clinical outcomes are limited. The purpose of this study was to investigate clinical outcomes of RT 106 infections compared with two other endemic strains of varying virulence. METHODS: This multicenter study evaluated adults hospitalized with C. difficile infection (CDI). C. difficile samples underwent PCR ribotyping and patients infected with RT 106 were compared to patients infected with a known hypervirulent strain (RT 027) and a strain associated with less virulence (RT 014-020). Electronic medical records were reviewed by blinded investigators to assess the primary outcome of poor clinical outcome (composite of initial clinical failure, discharge to a higher level of care, 90-day CDI recurrence, and CDI-contributable mortality). RESULTS: A total of 396 patients with CDI were identified (RT 106, 32.3%; RT 027, 29.3%; RT 014-020, 38.3%). Patients infected with RT 014-020 less often experienced a poor clinical outcome (40%) compared with RT 106 (56%) and RT 027 (65%) infection (P < 0.0001). After controlling for covariates and using RT 014-020 as a comparator, patients infected with RT 106 (OR, 2.25; 95% CI, 1.36-3.73) or RT 027 (OR, 2.56; 95% CI, 1.52-4.31) had higher odds of poor clinical outcome. Using RT 027 as the comparator, only RT 014-020 was associated with lower odds of poor clinical outcome (OR, 0.42; 95% CI, 0.27-0.65). CONCLUSION: This study demonstrated that the emergent C. difficile RT 106 was associated with increased rates of poor clinical outcomes compared to RT 014-020 and comparable poor clinical outcomes to RT 027. These findings can help to better understand the clinical significance of this and future emerging ribotypes.


Subject(s)
Clostridioides difficile/classification , Clostridioides difficile/genetics , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Cross Infection/epidemiology , Cross Infection/microbiology , Genotype , Aged , Aged, 80 and over , Bacterial Toxins/biosynthesis , Bacterial Toxins/genetics , Clostridioides difficile/isolation & purification , Female , Hospital Mortality , Hospitalization , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Ribotyping
6.
Anaerobe ; 69: 102352, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33640461

ABSTRACT

BACKGROUND: Fidaxomicin has novel pharmacologic effects on C. difficile spore formation including outgrowth inhibition and persistent spore attachment. However, the mechanism of fidaxomicin attachment on spores has not undergone rigorous microscopic studies. MATERIALS & METHODS: Fidaxomicin attachment to C. difficile spores of three distinct ribotypes and C. difficile mutant spores with inactivation of exosporium or spore-coat protein-coding genes were visualized using confocal microscopy with a fidaxomicin-bodipy compound (green fluorescence). The pharmacologic effect of the fidaxomicin-bodipy compound was determined. Confocal microscopy experiments included direct effect on C. difficile wild-type and mutant spores, effect of exosporium removal, and direct attachment to a comparator spore forming organism, Bacillus subtilis. RESULTS: The fidaxomicin-bodipy compound MIC was 1 mg/L compared to 0.06 mg/L for unlabeled fidaxomicin, a 16-fold increase. Using confocal microscopy, the intracellular localization of fidaxomicin into vegetative C. difficile cells was observed consistent with its RNA polymerase mechanism of action and inhibited spore outgrowth. The fidaxomicin-bodipy compound was visualized outside of the core of C. difficile spores with no co-localization with the membrane staining dye FM4-64. Exosporium removal reduced fidaxomicin-bodipy association with C. difficile spores. Reduced fidaxomicin-bodipy was observed in C. difficile mutant spores for the spore surface proteins CdeC and CotE. CONCLUSION: This study visualized a direct attachment of fidaxomicin to C. difficile spores that was diminished with mutants of specific exosporium and spore coat proteins. These data provide advanced insight regarding the anti-spore properties of fidaxomicin.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Cell Wall/drug effects , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Clostridium Infections/drug therapy , Fidaxomicin/therapeutic use , Spores, Bacterial/cytology , Spores, Bacterial/drug effects , Clostridioides difficile/cytology , Genetic Variation , Mutation , Ribotyping
7.
Anaerobe ; 70: 102387, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34044101

ABSTRACT

BACKGROUND: Fecal microbiota transplantation (FMT) aims to cure Clostridioides difficile infection (CDI) through reestablishing a healthy microbiome and restoring colonization resistance. Although often effective after one infusion, patients with continued microbiome disruptions may require multiple FMTs. In this N-of-1 study, we use a systems biology approach to evaluate CDI in a patient receiving chronic suppressive antibiotics with four failed FMTs over two years. METHODS: Seven stool samples were obtained between 2016-18 while the patient underwent five FMTs. Stool samples were cultured for C. difficile and underwent microbial characterization and functional gene analysis using shotgun metagenomics. C. difficile isolates were characterized through ribotyping, whole genome sequencing, metabolic pathway analysis, and minimum inhibitory concentration (MIC) determinations. RESULTS: Growing ten strains from each sample, the index and first four recurrent cultures were single strain ribotype F078-126, the fifth was a mixed culture of ribotypes F002 and F054, and the final culture was ribotype F002. One single nucleotide polymorphism (SNP) variant was identified in the RNA polymerase (RNAP) ß-subunit RpoB in the final isolated F078-126 strain when compared to previous F078-126 isolates. This SNV was associated with metabolic shifts but phenotypic differences in fidaxomicin MIC were not observed. Microbiome differences were observed over time during vancomycin therapy and after failed FMTs. CONCLUSION: This study highlights the importance of antimicrobial stewardship in patients receiving FMT. Continued antibiotics play a destructive role on a transplanted microbiome and applies selection pressure for resistance to the few antibiotics available to treat CDI.


Subject(s)
Clostridioides difficile/physiology , Clostridium Infections/therapy , Fecal Microbiota Transplantation , Anti-Bacterial Agents/administration & dosage , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Clostridioides difficile/drug effects , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Feces , Female , Gastrointestinal Microbiome/drug effects , Humans , Microbial Sensitivity Tests , Polymorphism, Single Nucleotide , Treatment Failure
8.
Article in English | MEDLINE | ID: mdl-32513796

ABSTRACT

Omadacycline is a potent aminomethylcycline with in vitro activity against Gram-positive, Gram-negative, and anaerobic bacteria. Preliminary data demonstrated that omadacycline has in vitro activity against Clostridioides difficile; however, large-scale in vitro studies have not been done. The purpose of this study was to assess the in vitro susceptibility of omadacycline and comparators on a large biobank of clinical C. difficile isolates. In vitroC. difficile susceptibility to omadacycline and comparators (fidaxomicin, metronidazole, and vancomycin) was assessed using the broth microdilution method. Minimum bactericidal concentrations (MBCs) and time-kill assays were assessed for pharmacodynamics analysis, and whole-genome sequencing was performed in a subset of isolates to assess distribution of MICs and resistance determinants. Two hundred fifty clinical C. difficile isolates collected between 2015 and 2018 were tested for in vitro susceptibility of omadacycline and comparators. Ribotypes included F001 (n = 5), F002 (n = 56), F014-020 (n = 66), F017 (n = 8), F027 (n = 53), F106 (n = 45), and F255 (n = 17). Omadacycline demonstrated potent in vitro activity with an MIC range of 0.016 to 0.13 µg/ml, an MIC50 of 0.031 µg/ml, and an MIC90 of 0.031 µg/ml. No difference was observed for omadacycline MIC50 and MIC90 values stratified by ribotype, disease severity, or vancomycin susceptibility. Bactericidal activity was confirmed in time-kill studies. No difference was observed in MIC based on C. difficile phylogeny. Further development of omadacycline as an intravenous and oral antibiotic directed toward C. difficile infection is warranted.


Subject(s)
Clostridioides difficile , Clostridioides , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Clostridioides difficile/genetics , Microbial Sensitivity Tests , Tetracyclines/pharmacology
9.
J Antimicrob Chemother ; 75(10): 2879-2884, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32719870

ABSTRACT

BACKGROUND: Eravacycline is a novel synthetic fluorocycline antibacterial approved for complicated intra-abdominal infections. OBJECTIVES: The purpose of this study was to assess the in vitro activities of eravacycline and comparator antibiotics against contemporary clinical isolates of Clostridioides difficile representing common ribotypes, including isolates with decreased susceptibility to metronidazole and vancomycin. METHODS: Clinical C. difficile strains from six common or emerging ribotypes were used to test the in vitro activities of eravacycline and comparator antibiotics (fidaxomicin, vancomycin and metronidazole) by broth microdilution. In addition, MBC experiments, time-kill kinetic studies and WGS experiments were performed. RESULTS: A total of 234 isolates were tested, including ribotypes RT001 (n = 37), RT002 (n = 41), RT014-020 (n = 39), RT027 (n = 42), RT106 (n = 38) and RT255 (n = 37). MIC50/90 values were lowest for eravacycline (≤0.0078/0.016 mg/L), followed by fidaxomicin (0.016/0.063 mg/L), metronidazole (0.25/1.0 mg/L) and vancomycin (2.0/4.0 mg/L). MBCs were lower for eravacycline compared with vancomycin for all ribotypes tested. Both vancomycin and eravacycline demonstrated bactericidal killing, including for epidemic RT027. The presence of the tetM or tetW resistance genes did not affect the MIC of eravacycline. CONCLUSIONS: This study demonstrated potent in vitro activity of eravacycline against a large collection of clinical C. difficile strains that was not affected by ribotype, susceptibility to vancomycin or the presence of certain tet resistance genes. Further development of eravacycline as an antibiotic to be used in patients with Clostridioides difficile infection is warranted.


Subject(s)
Clostridioides difficile , Clostridium Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Clostridioides , Clostridioides difficile/genetics , Clostridium Infections/drug therapy , Humans , Kinetics , Microbial Sensitivity Tests , Ribotyping , Tetracyclines
10.
J Antimicrob Chemother ; 75(12): 3635-3643, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32892222

ABSTRACT

BACKGROUND: Clostridioides difficile infection is the most common cause of healthcare-associated infections in the USA, with limited treatment options. Ibezapolstat is a novel DNA polymerase IIIC inhibitor with in vitro activity against C. difficile. OBJECTIVES AND METHODS: Randomized, double-blind, placebo-controlled study to assess the safety, tolerability and pharmacokinetics of ibezapolstat in healthy volunteers. Microbiome changes associated with ibezapolstat were compared with vancomycin over a 10 day course using shotgun metagenomics. RESULTS: A total of 62 subjects aged 31 ± 7 years (45% female; average BMI: 25 ± 3 kg/m2) were randomized. Ibezapolstat was well tolerated with a safety signal similar to placebo. Ibezapolstat had minimal systemic absorption with the majority of plasma concentrations less than 1 µg/mL. In the multiday, ascending dose study, ibezapolstat concentrations of 2000 µg/g of stool were observed by Day 2 and for the remainder of the dosing time period. In the multiday, multiple-dose arm, baseline microbiota was comparable between subjects that received ibezapolstat compared with vancomycin. At Day 10 of dosing, differential abundance analysis and ß-diversity demonstrated a distinct difference between the microbiome in subjects given vancomycin compared with either dose of ibezapolstat (P = 0.006). α-Diversity changes were characterized as an increase in the Actinobacteria phylum in subjects that received ibezapolstat and an increase in Proteobacteria in subjects given vancomycin. CONCLUSIONS: Ibezapolstat was shown to be safe and well tolerated, with minimal systemic exposure, high stool concentrations and a distinct microbiome profile compared with oral vancomycin. These results support further clinical development of ibezapolstat for patients with C. difficile infection.


Subject(s)
Clostridioides difficile , Clostridium Infections , Microbiota , Administration, Oral , Clostridium Infections/drug therapy , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Male
11.
Anaerobe ; 61: 102081, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31356958

ABSTRACT

Epidemiology of Clostridioides difficile (syn. Clostridium difficile) infection (CDI) in Bangladesh is poorly understood. This study assessed the epidemiology of CDI in hospitalized patients and hospital environmental contamination of toxigenic C. difficile at two large urban Bangladesh hospitals. This 12-month prospective observational cohort study collected stool samples from adults with diarrhea and recent antimicrobial exposure during 2017. Environmental samples were collected by swabbing surfaces of hospital common areas. Samples underwent toxigenic culture. C. difficile isolates were tested for toxins A and B and PCR-ribotyped. Of 208 stool samples, 18 (8.7%) were positive for toxigenic C. difficile. Of 400 environmental samples, 45 (11%) were positive for toxigenic C. difficile. Ribotypes present in ≥10% of stool isolates were 017 (38%), 053-163 (13%), and a novel ribotype (FP435 [13%]). Common ribotypes in environmental isolates were 017 (22%), 053-163 (11%), 106 (24%). This is the first report describing current epidemiology of CDI in at risk hospitalized adult patients in Bangladesh.


Subject(s)
Bacterial Toxins/genetics , Clostridioides difficile/classification , Clostridioides difficile/genetics , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Cross Infection , Hospitals , Adult , Bangladesh/epidemiology , Clostridioides difficile/isolation & purification , Female , Humans , Male , Middle Aged , Molecular Epidemiology , Public Health Surveillance , Ribotyping
12.
Clin Infect Dis ; 68(11): 1887-1893, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30204878

ABSTRACT

BACKGROUND: Clostridioides (formerly Clostridium) difficile infection (CDI) is associated with significant morbidity and mortality, including frequent hospitalizations. However, the impact of CDI after hospital discharge is poorly understood. The purpose of this study was to assess patient discharge disposition and understand CDI-related risk factors for nonhome discharge. METHODS: Using a nationally representative database of Veterans Health Administration (VHA) patients (2003-2014) and a validation database from hospitalized non-VHA patients in Houston, Texas, admission and discharge disposition was obtained for patients with CDI and matched controls. Incidence of and clinical/microbiologic risk factors for nonhome discharge were assessed using these databases. RESULTS: A total of 15173 VHA patients with CDI and 48599 non-CDI control patients originally admitted from the community were included. Significantly more patients with CDI were discharged to a nonhome location compared with controls (18% vs 8%; P < .0001), most commonly hospice/death (12%) or nursing home/long-term care facility (6%). Results were confirmed using a propensity-matched analysis and a validation cohort of 1941 hospitalized patients with CDI in Houston, Texas. Age, comorbidities, severe CDI, and ribotypes F027, F001, and F053-163 were associated with a nonhome discharge (P < .05 for all). CONCLUSIONS: Hospitalized patients with CDI frequently required a higher level of medical care residence at discharge compared with non-CDI patients. Risk factors for discharge to a higher level of care included CDI disease severity and variables associated with recurrent CDI.


Subject(s)
Clostridium Infections/complications , Clostridium Infections/etiology , Cross Infection/complications , Patient Discharge/statistics & numerical data , Aged , Cross Infection/microbiology , Databases, Factual , Enterocolitis, Pseudomembranous/etiology , Enterocolitis, Pseudomembranous/microbiology , Female , Humans , Incidence , Length of Stay , Male , Middle Aged , Multivariate Analysis , Propensity Score , Recurrence , Retrospective Studies , Risk Factors , Texas
13.
Article in English | MEDLINE | ID: mdl-31611365

ABSTRACT

Clostridioides difficile spores can survive in the environment in either mono- or mixed-species biofilms. However, no previous studies have investigated chemical disinfection of C. difficile spores embedded in biofilms. Thus, the purpose of this study was to assess the in vitro effectiveness of hospital disinfectants against C. difficile spores embedded within biofilms. Five unique C. difficile strains embedded in three different biofilm types grown for 72 or 120 h were exposed to seven different hospital disinfectants. C. difficile abundance [as log(number of CFU/milliliter)] was calculated after manufacturer-determined contact times along with biofilm biomass and microscopy. The primary analysis compared differences between C. difficile vegetative cell and spore counts as well as amounts of biomass after exposure to disinfectants. C. difficile vegetative cells and spores were recovered from biofilms regardless of the type of biofilm growth or biofilm growth time. No disinfectant was able to completely eliminate C. difficile from the biofilms. Overall, Clorox, ortho-phthalaldehyde (OPA), and Virex were most effective at killing C. difficile spores regardless of biofilm age, ribotype, or wash conditions (whether biofilms are washed or unwashed) (P = 0.001, each). Clorox and OPA were also effective at killing total vegetative cell growth (P = 0.001, each), but Virex was found to be ineffective against vegetative cell growth in biofilms (P = 0.77). Clorox and Virex were most effective in reducing biomass, followed by Nixall, OPA, and Vital Oxide. No disinfectant was able to completely eliminate C. difficile embedded within biofilms although differences among disinfectants were noted. Future research will be required to determine methods to eradicate this persister reservoir.


Subject(s)
Clostridioides difficile/drug effects , Disinfectants/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Biomass , Clostridioides difficile/growth & development , Clostridioides difficile/physiology , Clostridium Infections/prevention & control , Colony Count, Microbial , Cross Infection/prevention & control , Disease Reservoirs/microbiology , Disinfection/methods , Environmental Microbiology , Humans , In Vitro Techniques , Microbial Sensitivity Tests , Quaternary Ammonium Compounds/pharmacology , Ribotyping , Sodium Hypochlorite/pharmacology , Spores, Bacterial/drug effects , o-Phthalaldehyde/pharmacology
14.
Anaerobe ; 59: 107-111, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31207298

ABSTRACT

Animals such as domestic dogs and zoo animals reside in close proximity to humans and could contribute to the dissemination of Clostridioides difficile spores which are common in the community environment. The purpose of this study was to assess C. difficile colonization in domestic dogs attending a day boarding facility and zoo animals receiving systemic antibiotics. Stool samples and paw swabs were collected from dogs who attended a day boarding facility. Stool samples were also collected from zoo animals starting systemic antibiotics. Finally, environmental samples were collected from nearby public parks. Stool samples and swabs were incubated anaerobically in enrichment broth for C. difficile growth, PCR was done to confirm presence of toxin genes, and PCR ribotyping was performed for strain characterization. During the study period, 136 dog stool samples were obtained, the paws of 16 dogs were swabbed, and 250 environmental swabs from surrounding public parks were obtained. Twenty-three of 136 dog stool samples (17%) and 9 of 16 dog paws sampled (56%) grew toxigenic C. difficile. One hundred and four stool samples from 49 zoo animals were collected of which 19 (18%) grew toxigenic C. difficile. Rates of toxigenic C. difficile colonization increased significantly during antibiotic therapy (33%) and then returned to baseline during the follow-up (11%) period (p = 0.019). Fifty-five of 250 environmental swabs from public parks (22%) grew toxigenic C. difficile. Ribotypes associated with human disease including 106 and 014-020 were isolated from all sources. This study demonstrated a high rate of toxigenic C. difficile colonization in domestic dogs and zoo animals with ribotypes similar to those causing human disease. These results demonstrate the relationship between humans, animals, and the environment in the dissemination of spores.


Subject(s)
Animals, Zoo/microbiology , Clostridioides difficile/classification , Clostridioides difficile/genetics , Clostridium Infections/veterinary , Dog Diseases/epidemiology , Ribotyping , Animals , Bacteriological Techniques , Clostridioides difficile/isolation & purification , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Dog Diseases/microbiology , Dogs , Molecular Epidemiology , Polymerase Chain Reaction
15.
Anaerobe ; 56: 88-90, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30794875

ABSTRACT

Clostridioides difficile infection (CDI) is an emerging but often understudied infectious disease in developing countries. This study was aimed to isolate and characterize C. difficile from shoe sole swabs and diarrheal patient's stool samples in Bangladesh. We collected 94 shoe sole swabs samples from urban communities in Dhaka and 208 diarrheal stool samples from hospitalized patients over a period of 4 months. Samples were incubated anaerobically for C. difficile growth, confirmed toxigenic, and PCR-ribotyped. Eleven of 94 (11.7%) shoe sole swabs and 4 of 208 (1.9%) stool samples were culture positive of which 9 shoe sole isolates were toxigenic. Six PCR ribotypes from the 9 toxigenic isolates were identified with ribotype F014-020 being the most common (n = 4; 44%). The recently identified ribotype 106 strain was also identified. To the best of our knowledge, this is the first report of C. difficile culture, isolation and characterization from environmental sources in Bangladesh.


Subject(s)
Clostridioides difficile/classification , Clostridioides difficile/isolation & purification , Clostridium Infections/microbiology , Diarrhea/microbiology , Environmental Microbiology , Feces/microbiology , Ribotyping , Bacterial Toxins/genetics , Bangladesh , Clostridioides difficile/genetics , Humans , Polymerase Chain Reaction , Urban Population
16.
Ann Pharmacother ; 52(8): 754-762, 2018 08.
Article in English | MEDLINE | ID: mdl-29557208

ABSTRACT

BACKGROUND: The Accelerate PhenoTestTM BC kit (AXDX) provides rapid organism identification (ID) and antimicrobial susceptibility testing (AST) results. Its potential role for antimicrobial stewardship is unknown. OBJECTIVE: To compare the diagnostic accuracy of AXDX with conventional methods (CMs) and assess AXDX's potential role for antimicrobial stewardship in patients with Gram-positive bloodstream infections (BSIs). METHODS: This retrospective cohort study included adults with Staphylococcus aureus or Enterococcus spp BSIs from July 2014 to January 2016 at a tertiary care medical center. Available isolates were tested on AXDX, and ID and AST results from AXDX were compared with those from CMs (VITEK 2 or ETEST). The following antibiotics were assessed for categorical agreement (CA) and essential agreement (EA) between the methods: ampicillin and daptomycin ( Enterococcus spp only), erythromycin and cefoxitin ( S aureus only), linezolid, and vancomycin. Potential role of AXDX for stewardship was assessed via a retrospective audit by infectious diseases clinicians. RESULTS: We included 231 patients with S aureus (n = 112) or Enterococcus spp (n = 119) BSIs, and 106 unique isolates were available for ID and AST performance analyses. Sensitivity and specificity of AXDX for ID were 98.0% and 99.5%, respectively. CA and EA for the tested antibiotics were >97%. In Monte Carlo simulations, AXDX coupled with stewardship personnel (either 24/7 or Monday to Friday) would have allowed unnecessary therapy to be stopped and active/targeted therapy to be started ≥24 hours sooner in >50% of patients. CONCLUSIONS: Compared with CMs, AXDX had similar diagnostic accuracy and can potentially optimize therapy sooner in patients with Gram-positive BSIs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Stewardship , Gram-Positive Bacteria/drug effects , Gram-Positive Bacterial Infections/diagnosis , Microbial Sensitivity Tests , Adult , Gram-Positive Bacteria/isolation & purification , Gram-Positive Bacterial Infections/blood , Gram-Positive Bacterial Infections/microbiology , Humans , Retrospective Studies
17.
Curr Opin Gastroenterol ; 33(1): 1-7, 2017 01.
Article in English | MEDLINE | ID: mdl-28134686

ABSTRACT

PURPOSE OF REVIEW: Clostridium difficile infections (CDI) remain a challenge to treat clinically due primarily to limited number of antibiotics available and unacceptably high recurrence rates. Because of this, there has been significant demand for creating innovative therapeutics, which has resulted in the development of several novel antibiotics. RECENT FINDINGS: This review updates seven different antibiotics that are currently in development to treat CDI including fidaxomicin, surotomycin, ridinilazole, ramoplanin, cadazolid, LFF571, and CRS3123. Available preclinical and clinical data are compared between these antibiotics. SUMMARY: Many of these new antibiotics display almost ideal properties for antibiotics directed against CDI. Despite these properties, not all clinical development of these compounds has been successful. These studies have provided key insights into the pathogenesis of CDI and will continue to inform future drug development. Successful phase III clinical trials should result in several new and novel antibiotics to treat CDI.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Clostridium Infections/drug therapy , Benzimidazoles/therapeutic use , Benzopyrans/therapeutic use , Clinical Trials as Topic , Depsipeptides/therapeutic use , Humans , Lipopeptides/therapeutic use , Oxazolidinones/therapeutic use , Peptides, Cyclic/therapeutic use , Pyridines/therapeutic use , Thiazoles/therapeutic use , Thiophenes/therapeutic use
18.
Arch Microbiol ; 199(2): 317-323, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27730251

ABSTRACT

Molecular typing of Clostridium difficile is performed to assess strain relatedness or place strains within an epidemiological context. Different C. difficile ribotyping systems are available. However, a common strain library does not exist. We aimed to compare ribotyping results of 29 clinical C. difficile isolates by two methods: semiautomated PCR-ribotyping and fluorescent PCR-ribotyping. For certain ribotypes (n = 16/29; 55.2 %), the inter-laboratory reproducibility was consistent among multiple samples from individual subjects, while 54.8 % (n = 14/29) were discordant. Additionally, 11/29 ribotypes (38 %) and 12/29 ribotypes (41 %) did not match with known reference strains in the semiautomated PCR-fluorescent ribotyping systems' libraries, respectively. The identification of 027 ribotype by both systems was consistent for 75 % of the isolates. Discriminatory indices for the semiautomated PCR-ribotyping and fluorescent PCR-ribotyping systems are 0.906 and 0.886, respectively. Although ribotyping provides important epidemiologic insights, the lack of a common strain library makes interpretation of results using different ribotyping protocols difficult.


Subject(s)
Clostridioides difficile/classification , Polymerase Chain Reaction/methods , Ribotyping/methods , Clostridioides difficile/genetics , Clostridioides difficile/isolation & purification , Fluorescence , Humans , Reproducibility of Results
19.
Antimicrob Agents Chemother ; 60(6): 3519-23, 2016 06.
Article in English | MEDLINE | ID: mdl-27021314

ABSTRACT

Surotomycin is a cyclic lipopeptide in development for Clostridium difficile-associated diarrhea. This study aimed to assess the impact of surotomycin exposure on C. difficile toxin A and B concentrations and the associated changes in immune response in comparison to vancomycin and metronidazole. Time-kill curve assays were performed using strain R20291 (BI/NAP1/027) at supra-MICs (4× and 40×) and sub-MICs (0.5×) of surotomycin and comparators. Following treatment, CFU counts, toxin A and B concentrations, and cellular morphological changes using scanning electron microscopy were examined. Inflammatory response was determined by measuring interleukin-8 (IL-8) concentrations from polarized Caco-2 cells exposed to antibiotic-treated C. difficile growth media. Supra-MICs (4× and 40×) of surotomycin resulted in a reduction of vegetative cells over 72 h (4-log difference, P < 0.01) compared to controls. These results correlated with decreases of 77% and 68% in toxin A and B production at 48 h, respectively (P < 0.005, each), which resulted in a significant reduction in IL-8 concentration compared to controls. Similar results were observed with comparator antibiotics. Bacterial cell morphology showed that the cell wall was broken apart by surotomycin treatment at supra-MICs while sub-MIC studies showed a "deflated" phenotype plus a rippling effect. These results suggest that surotomycin has potent killing effects on C. difficile that results in reduced toxin production and attenuates the immune response similar to comparator antibiotics. The morphological data also confirm observations that surotomycin is a membrane-active antibiotic.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Bacterial Toxins/biosynthesis , Clostridioides difficile/drug effects , Clostridioides difficile/immunology , Enterotoxins/biosynthesis , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Caco-2 Cells , Cell Line, Tumor , Clostridioides difficile/metabolism , Humans , Interleukin-8/immunology , Metronidazole/pharmacology , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Vancomycin/pharmacology
20.
J Antimicrob Chemother ; 71(5): 1245-51, 2016 May.
Article in English | MEDLINE | ID: mdl-26895772

ABSTRACT

OBJECTIVES: Ridinilazole (SMT19969) is a narrow-spectrum, non-absorbable antimicrobial with activity against Clostridium difficile undergoing clinical trials. The purpose of this study was to assess the pharmacological activity of ridinilazole and assess the effects on cell morphology. METHODS: Antibiotic killing curves were performed using the epidemic C. difficile ribotype 027 strain, R20291, using supra-MIC (4× and 40×) and sub-MIC (0.125×, 0.25× and 0.5×) concentrations of ridinilazole. Following exposure, C. difficile cells were collected for cfu counts, toxin A and B production, and morphological changes using scanning electron and fluorescence microscopy. Human intestinal cells (Caco-2) were co-incubated with ridinilazole-treated C. difficile growth medium to determine the effects on host inflammatory response (IL-8). RESULTS: Treatment at supra-MIC concentrations (4× and 40× MIC) of ridinilazole resulted in a significant reduction in vegetative cells over 72 h (4 log difference, P < 0.01) compared with controls without inducing spore formation. These results correlated with a 75% decrease in toxin A production (P < 0.05) and a 96% decrease in toxin B production (P < 0.05). At sub-MIC levels (0.5× MIC), toxin A production was reduced by 91% (P < 0.01) and toxin B production was reduced by 100% (P < 0.001), which resulted in a 74% reduction in IL-8 release compared with controls (P < 0.05). Sub-MIC (0.5×)-treated cells formed filamentous structures ∼10-fold longer than control cells. Following fluorescence labelling, the cell septum was not forming in sub-MIC-treated cells, yet the DNA was dividing. CONCLUSIONS: Ridinilazole had robust killing effects on C. difficile that significantly reduced toxin production and attenuated the inflammatory response. Ridinilazole also elicited significant cell division effects suggesting a potential mechanism of action.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/metabolism , Benzimidazoles/pharmacology , Clostridioides difficile/drug effects , Pyridines/pharmacology , Caco-2 Cells , Clostridioides difficile/cytology , Clostridioides difficile/metabolism , Cytokines/metabolism , Epithelial Cells/microbiology , Humans , Microbial Sensitivity Tests , Microbial Viability/drug effects , Microscopy, Electron, Scanning
SELECTION OF CITATIONS
SEARCH DETAIL