Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
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.
Antimicrob Agents Chemother ; 60(2): 729-34, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26574013

ABSTRACT

S-649266 is a novel siderophore cephalosporin antibiotic with a catechol moiety on the 3-position side chain. Two sets of clinical isolate collections were used to evaluate the antimicrobial activity of S-649266 against Enterobacteriaceae. These sets included 617 global isolates collected between 2009 and 2011 and 233 ß-lactamase-identified isolates, including 47 KPC-, 49 NDM-, 12 VIM-, and 8 IMP-producers. The MIC90 values of S-649266 against the first set of Escherichia coli, Klebsiella pneumoniae, Serratia marcescens, Citrobacter freundii, Enterobacter aerogenes, and Enterobacter cloacae isolates were all ≤1 µg/ml, and there were only 8 isolates (1.3%) among these 617 clinical isolates with MIC values of ≥8 µg/ml. In the second set, the MIC values of S-649266 were ≤4 µg/ml against 109 strains among 116 KPC-producing and class B (metallo) carbapenemase-producing strains. In addition, S-649266 showed MIC values of ≤2 µg/ml against each of the 13 strains that produced other types of carbapenemases such as SME, NMC, and OXA-48. The mechanisms of the decreased susceptibility of 7 class B carbapenemase-producing strains with MIC values of ≥16 µg/ml are uncertain. This is the first report to demonstrate that S-649266, a novel siderophore cephalosporin, has significant antimicrobial activity against Enterobacteriaceae, including strains that produce carbapenemases such as KPC and NDM-1.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Enterobacteriaceae/drug effects , Siderophores/pharmacology , Bacterial Proteins/metabolism , Carbapenems/pharmacology , Enterobacteriaceae/isolation & purification , Escherichia coli Proteins/metabolism , Humans , Microbial Sensitivity Tests , beta-Lactamases/metabolism , Cefiderocol
5.
Antimicrob Agents Chemother ; 60(1): 180-9, 2016 01.
Article in English | MEDLINE | ID: mdl-26482300

ABSTRACT

GSK1322322 is a novel inhibitor of peptide deformylase (PDF) with good in vitro activity against bacteria associated with community-acquired pneumonia and skin infections. We have characterized the in vivo pharmacodynamics (PD) of GSK1322322 in immunocompetent animal models of infection with Streptococcus pneumoniae and Haemophilus influenzae (mouse lung model) and with Staphylococcus aureus (rat abscess model) and determined the pharmacokinetic (PK)/PD index that best correlates with efficacy and its magnitude. Oral PK studies with both models showed slightly higher-than-dose-proportional exposure, with 3-fold increases in area under the concentration-time curve (AUC) with doubling doses. GSK1322322 exhibited dose-dependent in vivo efficacy against multiple isolates of S. pneumoniae, H. influenzae, and S. aureus. Dose fractionation studies with two S. pneumoniae and S. aureus isolates showed that therapeutic outcome correlated best with the free AUC/MIC (fAUC/MIC) index in S. pneumoniae (R(2), 0.83), whereas fAUC/MIC and free maximum drug concentration (fCmax)/MIC were the best efficacy predictors for S. aureus (R(2), 0.9 and 0.91, respectively). Median daily fAUC/MIC values required for stasis and for a 1-log10 reduction in bacterial burden were 8.1 and 14.4 for 11 S. pneumoniae isolates (R(2), 0.62) and 7.2 and 13.0 for five H. influenzae isolates (R(2), 0.93). The data showed that for eight S. aureus isolates, fAUC correlated better with efficacy than fAUC/MIC (R(2), 0.91 and 0.76, respectively), as efficacious AUCs were similar for all isolates, independent of their GSK1322322 MIC (range, 0.5 to 4 µg/ml). Median fAUCs of 2.1 and 6.3 µg · h/ml were associated with stasis and 1-log10 reductions, respectively, for S. aureus.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Haemophilus Infections/drug therapy , Hydroxamic Acids/pharmacokinetics , Pneumonia, Pneumococcal/drug therapy , Staphylococcal Infections/drug therapy , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacology , Area Under Curve , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/blood , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacology , Haemophilus Infections/blood , Haemophilus Infections/microbiology , Haemophilus influenzae/drug effects , Haemophilus influenzae/enzymology , Haemophilus influenzae/growth & development , Hydroxamic Acids/blood , Hydroxamic Acids/pharmacology , Lung/drug effects , Lung/microbiology , Male , Mice , Microbial Sensitivity Tests , Pneumonia, Pneumococcal/blood , Pneumonia, Pneumococcal/microbiology , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Staphylococcus aureus/growth & development , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/growth & development
6.
J Antimicrob Chemother ; 71(3): 670-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26645269

ABSTRACT

OBJECTIVES: S-649266 is a parenteral siderophore cephalosporin antibiotic with a catechol moiety on its side chain. The in vitro antimicrobial activity of S-649266 against non-fermenting Gram-negative bacteria was evaluated and compared with the activities of meropenem, levofloxacin, cefepime, ceftazidime and piperacillin/tazobactam. METHODS: MIC values of S-649266 were determined in Mueller-Hinton broth or Iso-Sensitest broth supplemented with apo-transferrin. RESULTS: S-649266 showed potent in vitro activity against the non-fermenting Gram-negative bacteria Acinetobacter baumannii, Pseudomonas aeruginosa and Stenotrophomonas maltophilia, including MDR strains such as carbapenem-resistant A. baumannii and metallo-ß-lactamase-producing P. aeruginosa. MIC90s of S-649266 for A. baumannii, P. aeruginosa and S. maltophilia were 2, 1 and 0.5 mg/L, respectively, whereas MIC90s of meropenem were >16 mg/L. S-649266 showed potent in vitro activities against A. baumannii producing carbapenemases such as OXA-type ß-lactamases, and P. aeruginosa producing metallo-ß-lactamases such as IMP type and VIM type. MIC90 values for these A. baumannii strains and P. aeruginosa strains were 8 and 4 mg/L, respectively. CONCLUSIONS: S-649266 is a novel antibiotic with potent in vitro activity against a range of non-fermenting Gram-negative bacteria, including MDR strains.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Catechols/pharmacology , Cephalosporins/pharmacology , Pseudomonas aeruginosa/drug effects , Siderophores/pharmacology , Stenotrophomonas maltophilia/drug effects , Gram-Negative Bacteria/drug effects , Humans , Microbial Sensitivity Tests
7.
Antimicrob Agents Chemother ; 59(1): 289-98, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348524

ABSTRACT

GSK2251052, a novel leucyl-tRNA synthetase (LeuRS) inhibitor, was in development for the treatment of infections caused by multidrug-resistant Gram-negative pathogens. In a phase II study (study LRS114688) evaluating the efficacy of GSK2251052 in complicated urinary tract infections, resistance developed very rapidly in 3 of 14 subjects enrolled, with ≥32-fold increases in the GSK2251052 MIC of the infecting pathogen being detected. A fourth subject did not exhibit the development of resistance in the baseline pathogen but posttherapy did present with a different pathogen resistant to GSK2251052. Whole-genome DNA sequencing of Escherichia coli isolates collected longitudinally from two study LRS114688 subjects confirmed that GSK2251052 resistance was due to specific mutations, selected on the first day of therapy, in the LeuRS editing domain. Phylogenetic analysis strongly suggested that resistant Escherichia coli isolates resulted from clonal expansion of baseline susceptible strains. This resistance development likely resulted from the confluence of multiple factors, of which only some can be assessed preclinically. Our study shows the challenges of developing antibiotics and the importance of clinical studies to evaluate their effect on disease pathogenesis. (These studies have been registered at ClinicalTrials.gov under registration no. NCT01381549 for the study of complicated urinary tract infections and registration no. NCT01381562 for the study of complicated intra-abdominal infections.).


Subject(s)
Boron Compounds/pharmacology , Drug Resistance, Bacterial/drug effects , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Leucine-tRNA Ligase/antagonists & inhibitors , Urinary Tract Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents, Urinary/pharmacology , Boron Compounds/therapeutic use , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Genome, Bacterial , Humans , Mutation , Phylogeny , Urinary Tract Infections/microbiology
8.
Bioorg Med Chem Lett ; 21(24): 7483-8, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22030032

ABSTRACT

As part of our wider efforts to exploit novel mode of action antibacterials, we have discovered a series of cyclohexyl-amide compounds that has good Gram positive and Gram negative potency. The mechanism of action is via inhibition of bacterial topoisomerases II and IV. We have investigated various subunits in this series and report advanced studies on compound 7 which demonstrates good PK and in vivo efficacy properties.


Subject(s)
Amides/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , DNA Topoisomerases, Type II/chemistry , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Topoisomerase II Inhibitors/chemistry , Amides/chemical synthesis , Amides/pharmacokinetics , Animals , Anti-Bacterial Agents/chemical synthesis , Binding Sites , Computer Simulation , DNA Topoisomerases, Type II/metabolism , Dogs , Haplorhini , Humans , Microbial Sensitivity Tests , Protein Structure, Tertiary , Rats , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/pharmacokinetics
9.
Bioorg Med Chem Lett ; 21(24): 7489-95, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22047689

ABSTRACT

We have identified a series of amino-piperidine antibacterials with a good broad spectrum potency. We report the investigation of various subunits in this series and advanced studies on compound 8. Compound 8 possesses good pharmacokinetics, broad spectrum antibacterial activity and demonstrates oral efficacy in a rat lung infection model.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , DNA Topoisomerases, Type II/chemistry , Dioxanes/chemistry , Dioxanes/pharmacology , Naphthyridines/chemistry , Naphthyridines/pharmacology , Piperidines/chemistry , Topoisomerase II Inhibitors/chemistry , Animals , Anti-Bacterial Agents/therapeutic use , DNA Topoisomerases, Type II/metabolism , Dioxanes/therapeutic use , Disease Models, Animal , Dogs , Haplorhini , Humans , Lung Diseases/drug therapy , Microbial Sensitivity Tests , Naphthyridines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Rats , Structure-Activity Relationship , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use
10.
Front Immunol ; 12: 705360, 2021.
Article in English | MEDLINE | ID: mdl-34305945

ABSTRACT

Staphylococcus aureus is one of the most important human pathogens worldwide. Its high antibiotic resistance profile reinforces the need for new interventions like vaccines in addition to new antibiotics. Vaccine development efforts against S. aureus have failed so far however, the findings from these human clinical and non-clinical studies provide potential insight for such failures. Currently, research is focusing on identifying novel vaccine formulations able to elicit potent humoral and cellular immune responses. Translational science studies are attempting to discover correlates of protection using animal models as well as in vitro and ex vivo models assessing efficacy of vaccine candidates. Several new vaccine candidates are being tested in human clinical trials in a variety of target populations. In addition to vaccines, bacteriophages, monoclonal antibodies, centyrins and new classes of antibiotics are being developed. Some of these have been tested in humans with encouraging results. The complexity of the diseases and the range of the target populations affected by this pathogen will require a multipronged approach using different interventions, which will be discussed in this review.


Subject(s)
Staphylococcal Infections/prevention & control , Staphylococcal Vaccines , Staphylococcus aureus/immunology , Vaccine Development , Adjuvants, Immunologic , Animals , Antigens, Bacterial/immunology , Clinical Trials as Topic , Extracellular Vesicles/immunology , Glycoconjugates/immunology , Gram-Negative Bacteria/immunology , Host-Pathogen Interactions , Humans , Immunity, Cellular , Immunity, Humoral , Immunogenicity, Vaccine , In Vitro Techniques , Mice , Models, Animal , Nucleic Acid-Based Vaccines/immunology , Periplasm/immunology , Recombinant Proteins/immunology , Staphylococcal Vaccines/immunology , Staphylococcal Vaccines/therapeutic use , Translational Science, Biomedical , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology
12.
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
13.
J Med Chem ; 45(10): 1959-62, 2002 May 09.
Article in English | MEDLINE | ID: mdl-11985462

ABSTRACT

Potent nanomolar inhibitors of Staphylococcus aureus methionyl tRNA synthetase have been derived from a file compound high throughput screening hit. Optimized compounds show excellent antibacterial activity against staphylococcal and enterococcal pathogens, including strains resistant to clinical antibiotics. Compound 11 demonstrated in vivo efficacy in an S. aureus rat abscess infection model.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Enterococcus/drug effects , Enzyme Inhibitors/chemical synthesis , Methionine-tRNA Ligase/antagonists & inhibitors , Quinolones/chemical synthesis , Staphylococcus/drug effects , Abscess/drug therapy , Abscess/microbiology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Quinolones/chemistry , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Structure-Activity Relationship
14.
J Med Chem ; 46(9): 1627-35, 2003 Apr 24.
Article in English | MEDLINE | ID: mdl-12699381

ABSTRACT

Bacterial enoyl-ACP reductase (FabI) is responsible for catalyzing the final step of bacterial fatty acid biosynthesis and is an attractive target for the development of novel antibacterial agents. Previously we reported the development of FabI inhibitor 4 with narrow spectrum antimicrobial activity and in vivo efficacy against Staphylococcus aureus via intraperitoneal (ip) administration. Through iterative medicinal chemistry aided by X-ray crystal structure analysis, a new series of inhibitors has been developed with greatly increased potency against FabI-containing organisms. Several of these new inhibitors have potent antibacterial activity against multidrug resistant strains of S. aureus, and compound 30 demonstrates exceptional oral (po) in vivo efficacy in a S. aureus infection model in rats. While optimizing FabI inhibitory activity, compounds 29 and 30 were identified as having low micromolar FabK inhibitory activity, thereby increasing the antimicrobial spectrum of these compounds to include the FabK-containing pathogens Streptococcus pneumoniae and Enterococcus faecalis. The results described herein support the hypothesis that bacterial enoyl-ACP reductases are valid targets for antibacterial agents.


Subject(s)
Acrylamides/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Fatty Acid Synthases/antagonists & inhibitors , Indoles/chemical synthesis , Naphthyridines/chemical synthesis , Oxidoreductases/antagonists & inhibitors , Abscess/drug therapy , Acrylamides/chemistry , Acrylamides/pharmacology , Administration, Oral , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , Drug Resistance, Bacterial , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Enterococcus faecalis/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Haemophilus influenzae/drug effects , Indoles/chemistry , Indoles/pharmacology , Microbial Sensitivity Tests , Models, Molecular , Naphthyridines/chemistry , Naphthyridines/pharmacology , Rats , Staphylococcus aureus/drug effects , Stereoisomerism , Structure-Activity Relationship , Triclosan/pharmacology
15.
J Med Chem ; 45(15): 3246-56, 2002 Jul 18.
Article in English | MEDLINE | ID: mdl-12109908

ABSTRACT

Bacterial enoyl-ACP reductase (FabI) catalyzes the final step in each cycle of bacterial fatty acid biosynthesis and is an attractive target for the development of new antibacterial agents. Our efforts to identify potent, selective FabI inhibitors began with screening of the GlaxoSmithKline proprietary compound collection, which identified several small-molecule inhibitors of Staphylococcus aureus FabI. Through a combination of iterative medicinal chemistry and X-ray crystal structure based design, one of these leads was developed into the novel aminopyridine derivative 9, a low micromolar inhibitor of FabI from S. aureus (IC(50) = 2.4 microM) and Haemophilus influenzae (IC(50) = 4.2 microM). Compound 9 has good in vitro antibacterial activity against several organisms, including S. aureus (MIC = 0.5 microg/mL), and is effective in vivo in a S. aureus groin abscess infection model in rats. Through FabI overexpressor and macromolecular synthesis studies, the mode of action of 9 has been confirmed to be inhibition of fatty acid biosynthesis via inhibition of FabI. Taken together, these results support FabI as a valid antibacterial target and demonstrate the potential of small-molecule FabI inhibitors for the treatment of bacterial infections.


Subject(s)
Acrylamides/chemical synthesis , Aminopyridines/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Fatty Acid Synthases/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Acrylamides/chemistry , Acrylamides/pharmacology , Aminopyridines/chemistry , Aminopyridines/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , Databases, Factual , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/chemistry , Haemophilus influenzae/drug effects , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Models, Molecular , Oxidoreductases/chemistry , Rats , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Structure-Activity Relationship
16.
Ann N Y Acad Sci ; 1213: 5-19, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21058956

ABSTRACT

The discovery of novel antibiotic classes has not kept pace with the growing threat of bacterial resistance. Antibiotic candidates that act at new targets or via distinct mechanisms have the greatest potential to overcome resistance; however, novel approaches are also associated with higher attrition and longer timelines. This uncertainty has contributed to the withdrawal from antibiotic programs by many pharmaceutical companies. Genomic approaches have not yielded satisfactory results, in part due to nascent knowledge about unprecedented molecular targets, the challenge of achieving antibacterial activity by lead optimization of enzyme inhibitors, and the limitations of compound screening libraries for antibacterial discovery. Enhanced diversity of compound screening banks, entry into new chemical space, and new screening technologies are currently being exploited to improve hit rates for antibacterial discovery. Antibacterial compound lead optimization faces hurdles associated with the high plasma exposures required for efficacy. Lead optimization would be enhanced by the identification of new antibiotic classes with improved tractability and by expanding the predictability of in vitro safety assays. Implementing multiple screening and target identification strategies is recommended for improving the likelihood of discovering new antibacterial compounds that address unmet needs.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Drug Discovery/methods , Drug Resistance, Bacterial/drug effects , Animals , Drug Design , Drug Evaluation, Preclinical/methods , Drug Industry/methods , Humans , Microbial Sensitivity Tests
17.
J Microbiol Methods ; 83(2): 254-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20801170

ABSTRACT

We developed a homogenous microtiter based assay using the cationic dye 3, 3'-Diethyloxacarbocyanine iodide, DiOC2(3), to measure the effect of compounds on membrane potential in Staphylococcus aureus. In a screen of 372 compounds from a synthetic compound collection with anti-Escherichia coli activity due to unknown modes of action at least 17% demonstrated potent membrane activity, enabling rapid discrimination of nuisance compounds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriological Techniques/methods , High-Throughput Screening Assays/methods , Membrane Potentials/drug effects , Staphylococcus aureus/drug effects , Carbocyanines/metabolism , Escherichia coli/drug effects , Inhibitory Concentration 50
18.
Antimicrob Agents Chemother ; 51(6): 2048-52, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17404009

ABSTRACT

To assess their effects on susceptibility to retapamulin in Staphylococcus aureus, first-, second-, and third-step mutants with elevated MICs to tiamulin and other investigational pleuromutilin compounds were isolated and characterized through exposure to high drug concentrations. All first- and second-step mutations were in rplC, encoding ribosomal protein L3. Most third-step mutants acquired a third mutation in rplC. While first- and second-step mutations did cause an elevation in tiamulin and retapamulin MICs, a significant decrease in activity was not seen until a third mutation was acquired. All third-step mutants exhibited severe growth defects, and faster-growing variants arose at a high frequency from most isolates. These faster-growing variants were found to be more susceptible to pleuromutilins. In the case of a mutant with three alterations in rplC, the fast-growing variants acquired an additional mutation in rplC. In the case of fast-growing variants of isolates with two mutations in rplC and at least one mutation at an unmapped locus, one of the two rplC mutations reverted to wild type. These data indicate that mutations in rplC that lead to pleuromutilin resistance have a direct, negative effect on fitness. While reduction in activity of retapamulin against S. aureus can be seen through mutations in rplC, it is likely that target-specific resistance to retapamulin will be slow to emerge due to the need for three mutations for a significant effect on activity and the fitness cost of each mutational step.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Mutation , Ribosomal Proteins/genetics , Staphylococcus aureus/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Diterpenes/pharmacology , Humans , Microbial Sensitivity Tests , Polycyclic Compounds , Ribosomal Protein L3 , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Pleuromutilins
20.
Antimicrob Agents Chemother ; 50(11): 3882-5, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17065625

ABSTRACT

The in vitro activity of retapamulin was determined and compared to that of topical and community antibiotics. The MIC(90)s of retapamulin against Staphylococcus aureus and Streptococcus pyogenes were 0.12 microg/ml and 0.016 microg/ml, respectively. Retapamulin has a low propensity to select resistance and produces an in vitro postantibiotic effect.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Bacteria/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Diterpenes/pharmacology , Drug Resistance, Bacterial , Haemophilus influenzae/drug effects , Humans , Microbial Sensitivity Tests , Moraxella catarrhalis/drug effects , Mupirocin/pharmacology , Polycyclic Compounds , Skin Diseases, Bacterial/microbiology , Staphylococcus/drug effects , Streptococcus/drug effects , Pleuromutilins
SELECTION OF CITATIONS
SEARCH DETAIL