Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-31061153

ABSTRACT

Gepotidacin is a first-in-class triazaacenaphthylene antibacterial that inhibits bacterial type II topoisomerases and has in vitro activity against a range of bacterial pathogens, including Escherichia coli Urinary tract infections often progress to pyelonephritis and are a worldwide problem due to the prevalence of multidrug-resistant E. coli strains. This study evaluated the in vivo efficacy of gepotidacin against four strains of multidrug-resistant E. coli in a rat pyelonephritis model. Infected rats received controlled intravenous infusions of gepotidacin every 12 h for 4 days that recreated human systemic exposures from oral gepotidacin (800 or 1,500 mg twice daily for 4 days). Liquid chromatography-tandem mass spectrometry analysis of blood samples and kidney homogenates showed that gepotidacin levels were 6- to 7-fold higher in kidneys than in blood. Across experiments with 4-day gepotidacin treatments, bacterial CFU in kidneys were reduced by 2.9 to 4.9 log10 compared to pretreatment levels, and bladder CFU were reduced to the lower limit of detection (1.2 log10). The efficacies of 800- and 1,500-mg gepotidacin exposures were statistically similar. A time-course experiment indicated that a period of more than 24 h of gepotidacin treatment was required for efficacy and that 4 days were needed for maximal response. Overall, these results demonstrate that the recreated human exposures of gepotidacin studied were effective in an animal model of pyelonephritis caused by multidrug-resistant E. coli and that further evaluation for clinical use is warranted.


Subject(s)
Acenaphthenes/therapeutic use , Anti-Bacterial Agents/therapeutic use , Escherichia coli/drug effects , Escherichia coli/pathogenicity , Heterocyclic Compounds, 3-Ring/therapeutic use , Pyelonephritis/drug therapy , Animals , Humans , Kidney/drug effects , Kidney/metabolism , Male , Microbial Sensitivity Tests , Pyelonephritis/microbiology , Rats , Rats, Sprague-Dawley
2.
Article in English | MEDLINE | ID: mdl-28807913

ABSTRACT

Directly testing proposed clinical dosing regimens in nonclinical studies can reduce the risk during the development of novel antibacterial agents. Optimal dosing regimens can be identified in animal models by testing recreated human pharmacokinetic profiles. An example of this approach using continuous intravenous infusions of GSK1322322 in immunocompetent rats to evaluate recreated human exposures from phase I trials in pneumonia models with Streptococcus pneumoniae and Haemophilus influenzae and an abscess model with Staphylococcus aureus is presented. GSK1322322 was administered via continuous intravenous infusion to recreate 1,000- or 1,500-mg oral doses every 12 h in humans. Significant reductions (P ≤ 0.05 for all comparisons) in bacterial numbers compared with those for the baseline controls were observed for S. pneumoniae and H. influenzae (mean log10 reductions, 1.6 to ≥2.7 and 1.8 to 3.3 CFU/lungs, respectively) with the recreated 1,000-mg oral dose. This profile was also efficacious against S. aureus (mean log10 reduction, 1.9 to 2.4 CFU/abscess). There was a nonsignificant trend for improved efficacy against S. aureus with the 1,500-mg oral dose (mean log10 reduction, 2.4 to 3.1 CFU/abscess). These results demonstrate that the human oral 1,000- or 1,500-mg exposure profiles of GSK1322322 recreated in rats were effective against representative community-associated pathogens and supported selection of the 1,500-mg oral dose given every 12 h for a phase II clinical skin infection study. Furthermore, this work exemplifies how the testing of recreated human pharmacokinetic profiles can be incorporated into the development process and serve as an aid for selecting optimal dosing regimens prior to conducting large-scale clinical studies.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Haemophilus influenzae/drug effects , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/therapeutic use , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Animals , Anti-Bacterial Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Disease Models, Animal , Drug Administration Schedule , Haemophilus Infections/drug therapy , Humans , Hydroxamic Acids/pharmacokinetics , Male , Microbial Sensitivity Tests , Pneumococcal Infections/drug therapy , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/drug therapy
3.
Article in English | MEDLINE | ID: mdl-28630178

ABSTRACT

Cefiderocol (S-649266), a novel siderophore cephalosporin, shows potent activity against carbapenem-resistant Gram-negative bacilli. In this study, we evaluated the efficacy of cefiderocol against carbapenem-resistant Gram-negative bacilli (Pseudomonas aeruginosa, Acinetobacter baumannii, and Klebsiella pneumoniae) in immunocompetent-rat respiratory tract infection models recreating plasma pharmacokinetics (PK) profiles in healthy human subjects. A total of 6 clinical isolates (1 cephalosporin-susceptible P. aeruginosa isolate, 1 multidrug-resistant P. aeruginosa isolate, 2 multidrug-resistant A. baumannii isolates, and 2 carbapenem-resistant K. pneumoniae isolates) were evaluated. Four-day treatment with a human exposure of 1 g ceftazidime every 8 h as a 0.5-h infusion showed potent efficacy only against a ceftazidime-susceptible isolate, not against five ceftazidime-resistant isolates harboring carbapenemase. With cefiderocol, a human exposure of 2 g every 8 h as a 3-h infusion for 4 days produced a >3 log10 reduction in the number of viable cells of these carbapenem-resistant isolates in the lungs. When the infusion time was 1 h, bactericidal activity was also observed against all isolates tested, although for 2 of 5 carbapenem-resistant isolates, a 3 log10 reduction was not achieved. The difference in efficacy achieved by changing the infusion period from 1 h to 3 h was considered to be due to the higher percentage of the dosing interval during which free-drug concentrations were above the MIC (%fTMIC), as observed for ß-lactam antibiotics. These results suggest the potential utility of cefiderocol for the treatment of lung infections caused by carbapenem-resistant P. aeruginosa, A. baumannii, and K. pneumoniae strains.


Subject(s)
Acinetobacter Infections/drug therapy , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/therapeutic use , Cephalosporins/pharmacokinetics , Cephalosporins/therapeutic use , Klebsiella Infections/drug therapy , Pseudomonas Infections/drug therapy , Respiratory Tract Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Animals , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/genetics , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Male , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Rats , Rats, Sprague-Dawley , Respiratory Tract Infections/microbiology , beta-Lactam Resistance/genetics , Cefiderocol
4.
J Vis Exp ; (119)2017 01 02.
Article in English | MEDLINE | ID: mdl-28117818

ABSTRACT

Efficacy of candidate antibacterial treatments must be demonstrated in animal models of infection as part of the discovery and development process, preferably in models which mimic the intended clinical indication. A method for inducing robust lung infections in immunocompetent rats and mice is described which allows for the assessment of treatments in a model of serious pneumonia caused by S. pneumoniae, H. influenzae, P. aeruginosa, K. pneumoniae or A. baumannii. Animals are anesthetized, and an agar-based inoculum is deposited deep into the lung via nonsurgical intratracheal intubation. The resulting infection is consistent, reproducible, and stable for at least 48 h and up to 96 h for most isolates. Studies with marketed antibacterials have demonstrated good correlation between in vivo efficacy and in vitro susceptibility, and concordance between pharmacokinetic/pharmacodynamic targets determined in this model and clinically accepted targets has been observed. Although there is an initial time investment when learning the technique, it can be performed quickly and efficiently once proficiency is achieved. Benefits of the model include elimination of the neutropenic requirement, increased robustness and reproducibility, ability to study more pathogens and isolates, improved flexibility in study design and establishment of a challenging infection in an immunocompetent host.


Subject(s)
Anti-Bacterial Agents/pharmacology , Pneumonia, Bacterial/drug therapy , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/pathogenicity , Animals , Disease Models, Animal , Haemophilus Infections/drug therapy , Haemophilus influenzae/pathogenicity , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/pathogenicity , Male , Mice , Pneumonia, Bacterial/microbiology , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/microbiology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Rats, Sprague-Dawley , Reproducibility of Results , Streptococcus pneumoniae/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...