Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
J Med Chem ; 63(20): 11882-11901, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32914979

ABSTRACT

Herein, we report spiropyrimidinetriones (SPTs) incorporating N-linked azole substituents on a benzisoxazole scaffold with improved Gram-positive antibacterial activity relative to previously described analogues. SPTs have an unusual spirocyclic architecture and represent a new antibacterial class of bacterial DNA gyrase and topoisomerase IV inhibitors. They are not cross-resistant to fluoroquinolones and other DNA gyrase/topoisomerase IV inhibitors used clinically. The activity of the SPTs was assessed for DNA gyrase inhibition, and the antibacterial activity across Gram-positive and Gram-negative pathogens with N-linked 1,2,4-triazoles substituted on the 5-position provides the most worthwhile profile. Directed nucleophilic and electrophilic chemistry was developed to vary this 5-position with carbon, nitrogen, or oxygen substituents and explore structure-activity relationships including those around a target binding model. Compounds with favorable pharmacokinetic parameters were identified, and two compounds demonstrated cidality in a mouse model of Staphylococcus aureus infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA Gyrase/metabolism , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Topoisomerase II Inhibitors/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Azoles/chemistry , Azoles/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Isoxazoles/chemistry , Isoxazoles/pharmacology , Mice , Microbial Sensitivity Tests , Molecular Structure , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Rats , Rats, Wistar , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry
2.
Antimicrob Agents Chemother ; 59(12): 7743-52, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26438502

ABSTRACT

The objective of this study was to investigate the risk of attenuated efficacy due to adaptive resistance for the siderophore-conjugated monocarbam SMC-3176 in Pseudomonas aeruginosa by using a pharmacokinetic/pharmacodynamic (PK/PD) approach. MICs were determined in cation-adjusted Mueller-Hinton broth (MHB) and in Chelex-treated, dialyzed MHB (CDMHB). Spontaneous resistance was assessed at 2× to 16× the MIC and the resulting mutants sequenced. Efficacy was evaluated in a neutropenic mouse thigh model at 3.13 to 400 mg/kg of body weight every 3 h for 24 h and analyzed for association with free time above the MIC (fT>MIC). To closer emulate the conditions of the in vivo model, we developed a novel assay testing activity mouse whole blood (WB). All mutations were found in genes related to iron uptake: piuA, piuC, pirR, fecI, and pvdS. Against four P. aeruginosa isolates, SMC-3176 displayed predictable efficacy corresponding to the fT>MIC using the MIC in CDMHB (R(2) = 0.968 to 0.985), with stasis to 2-log kill achieved at 59.4 to 81.1%. Efficacy did not translate for P. aeruginosa isolate JJ 4-36, as the in vivo responses were inconsistent with fT>MIC exposures and implied a threshold concentration that was greater than the MIC. The results of the mouse WB assay indicated that efficacy was not predictable using the MIC for JJ 4-36 and four additional isolates, against which in vivo failures of another siderophore-conjugated ß-lactam were previously reported. SMC-3176 carries a risk of attenuated efficacy in P. aeruginosa due to rapid adaptive resistance preventing entry via the siderophore-mediated iron uptake systems. Substantial in vivo testing is warranted for compounds using the siderophore approach to thoroughly screen for this in vitro-in vivo disconnect in P. aeruginosa.


Subject(s)
Anti-Bacterial Agents/pharmacology , Azetidines/pharmacology , Drug Resistance, Bacterial/genetics , Pseudomonas aeruginosa/metabolism , Siderophores/pharmacology , Sulfonamides/pharmacology , Animals , Anti-Bacterial Agents/pharmacokinetics , Azetidines/pharmacokinetics , Female , Iron/metabolism , Mice , Mice, Inbred ICR , Microbial Sensitivity Tests , Oligopeptides/metabolism , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Siderophores/pharmacokinetics , Sulfonamides/pharmacokinetics , beta-Lactamases/metabolism
3.
Bioorg Med Chem Lett ; 25(22): 5172-7, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26463129

ABSTRACT

Two novel compounds, pyridopyrimidines (1) and naphthyridines (2) were identified as potent inhibitors of bacterial NAD(+)-dependent DNA ligase (Lig) A in a fragment screening. SAR was guided by molecular modeling and X-ray crystallography. It was observed that the diaminonitrile pharmacophore made a key interaction with the ligase enzyme, specifically residues Glu114, Lys291, and Leu117. Synthetic challenges limited opportunities for diversification of the naphthyridine core, therefore most of the SAR was focused on a pyridopyrimidine scaffold. The initial diversification at R(1) improved both enzyme and cell potency. Further SAR developed at the R(2) position using the Negishi cross-coupling reaction provided several compounds, among these compounds 22g showed good enzyme potency and cellular potency.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , DNA Ligases/antagonists & inhibitors , NAD/metabolism , Naphthyridines/pharmacology , Pyrimidines/pharmacology , Anti-Bacterial Agents/chemical synthesis , Bacterial Proteins/chemistry , DNA Ligases/chemistry , Haemophilus influenzae/drug effects , Microbial Sensitivity Tests , Naphthyridines/chemical synthesis , Pyrimidines/chemical synthesis , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Structure-Activity Relationship
5.
Sci Rep ; 5: 11827, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26168713

ABSTRACT

With the diminishing effectiveness of current antibacterial therapies, it is critically important to discover agents that operate by a mechanism that circumvents existing resistance. ETX0914, the first of a new class of antibacterial agent targeted for the treatment of gonorrhea, operates by a novel mode-of-inhibition against bacterial type II topoisomerases. Incorporating an oxazolidinone on the scaffold mitigated toxicological issues often seen with topoisomerase inhibitors. Organisms resistant to other topoisomerase inhibitors were not cross-resistant with ETX0914 nor were spontaneous resistant mutants to ETX0914 cross-resistant with other topoisomerase inhibitor classes, including the widely used fluoroquinolone class. Preclinical evaluation of ETX0914 pharmacokinetics and pharmacodynamics showed distribution into vascular tissues and efficacy in a murine Staphylococcus aureus infection model that served as a surrogate for predicting efficacious exposures for the treatment of Neisseria gonorrhoeae infections. A wide safety margin to the efficacious exposure in toxicological evaluations supported progression to Phase 1. Dosing ETX0914 in human volunteers showed sufficient exposure and minimal adverse effects to expect a highly efficacious anti-gonorrhea therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Barbiturates/pharmacology , Barbiturates/therapeutic use , Gonorrhea/drug therapy , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Adult , Animals , Anti-Bacterial Agents/chemistry , Barbiturates/chemistry , DNA Topoisomerases, Type II/chemistry , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Resistance, Bacterial , Female , Fluoroquinolones/pharmacology , Gonorrhea/microbiology , Haplorhini , Humans , Isoxazoles , Male , Mice , Microbial Sensitivity Tests , Middle Aged , Models, Molecular , Molecular Conformation , Morpholines , Mutation , Neisseria gonorrhoeae/drug effects , Neisseria gonorrhoeae/genetics , Oxazolidinones , Rats , Spiro Compounds/chemistry , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Topoisomerase II Inhibitors/chemistry , Young Adult
6.
J Med Chem ; 58(15): 6264-82, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26158756

ABSTRACT

A novel class of bacterial type-II topoisomerase inhibitor displaying a spiropyrimidinetrione architecture fused to a benzisoxazole scaffold shows potent activity against Gram-positive and fastidious Gram-negative bacteria. Here, we describe a series of N-linked oxazolidinone substituents on the benzisoxazole that improve upon the antibacterial activity of initially described compounds of the class, show favorable PK properties, and demonstrate efficacy in an in vivo Staphylococcus aureus infection model. Inhibition of the topoisomerases DNA gyrase and topoisomerase IV from both Gram-positive and a Gram-negative organisms was demonstrated. Compounds showed a clean in vitro toxicity profile, including no genotoxicity and no bone marrow toxicity at the highest evaluated concentrations or other issues that have been problematic for some fluoroquinolones. Compound 1u was identified for advancement into human clinical trials for treatment of uncomplicated gonorrhea based on a variety of beneficial attributes including the potent activity and the favorable safety profile.


Subject(s)
Isoxazoles/chemistry , Oxazolidinones/chemistry , Pyrimidines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Cell Line , Crystallography, X-Ray , Dogs , Drug Discovery , Humans , Male , Pyrimidines/chemistry , Pyrimidines/toxicity , Rats , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/toxicity
7.
Antimicrob Agents Chemother ; 59(9): 5278-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26077256

ABSTRACT

The type II topoisomerases DNA gyrase and topoisomerase IV are clinically validated bacterial targets that catalyze the modulation of DNA topology that is vital to DNA replication, repair, and decatenation. Increasing resistance to fluoroquinolones, which trap the topoisomerase-DNA complex, has led to significant efforts in the discovery of novel inhibitors of these targets. AZ6142 is a member of the class of novel bacterial topoisomerase inhibitors (NBTIs) that utilizes a distinct mechanism to trap the protein-DNA complex. AZ6142 has very potent activity against Gram-positive organisms, including Staphylococcus aureus, Streptococcus pneumoniae, and Streptococcus pyogenes. In this study, we determined the frequencies of resistance to AZ6142 and other representative NBTI compounds in S. aureus and S. pneumoniae. The frequencies of selection of resistant mutants at 4× the MIC were 1.7 × 10(-8) for S. aureus and <5.5 × 10(-10) for S. pneumoniae. To improve our understanding of the NBTI mechanism of inhibition, the resistant S. aureus mutants were characterized and 20 unique substitutions in the topoisomerase subunits were identified. Many of these substitutions were located outside the NBTI binding pocket and impact the susceptibility of AZ6142, resulting in a 4- to 32-fold elevation in the MIC over the wild-type parent strain. Data on cross-resistance with other NBTIs and fluoroquinolones enabled the differentiation of scaffold-specific changes from compound-specific variations. Our results suggest that AZ6142 inhibits both type II topoisomerases in S. aureus but that DNA gyrase is the primary target. Further, the genotype of the resistant mutants suggests that domain conformations and DNA interactions may uniquely impact NBTIs compared to fluoroquinolones.


Subject(s)
Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Topoisomerase Inhibitors/pharmacology , DNA Gyrase/metabolism , DNA Topoisomerases, Type II/metabolism , Fluoroquinolones/pharmacology , Microbial Sensitivity Tests , Staphylococcus aureus/enzymology
8.
Antimicrob Agents Chemother ; 59(4): 2337-42, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25645840

ABSTRACT

A pyridodiazepine amine inhibitor of Helicobacter pylori glutamate racemase (MurI) was characterized. The compound was selectively active against H. pylori, and growth suppression was shown to be mediated through the inhibition of MurI by several methods. In killing kinetics experiments, the compound showed concentration-independent activity, with about a 2-log loss of viability in 24 h. A demonstration of efficacy in a mouse infection model was attempted but not achieved, and this was attributed to the failure to attain extended exposure levels above the MIC for >95% of the time. This index and magnitude were derived from pharmacokinetic-pharmacodynamic (PK-PD) studies with amoxicillin, another inhibitor of peptidoglycan biosynthesis that showed slow killing kinetics similar to those of the pyridodiazepine amines. These studies indicate that MurI and other enzymes involved in peptidoglycan biosynthesis may be less desirable targets for monotherapy directed against H. pylori if once-a-day dosing is required.


Subject(s)
Amino Acid Isomerases/antagonists & inhibitors , Anti-Bacterial Agents/therapeutic use , Azepines/therapeutic use , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Helicobacter pylori/enzymology , Pyridines/therapeutic use , Amoxicillin/pharmacokinetics , Amoxicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacokinetics , Azepines/pharmacokinetics , Female , Gastric Mucosa/metabolism , Helicobacter Infections/blood , Helicobacter Infections/microbiology , Kinetics , Mice , Microbial Sensitivity Tests , Peptidoglycan/metabolism , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley
9.
Antimicrob Agents Chemother ; 59(3): 1478-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534723

ABSTRACT

The unmet medical need for novel intervention strategies to treat Neisseria gonorrhoeae infections is significant and increasing, as rapidly emerging resistance in this pathogen is threatening to eliminate the currently available treatment options. AZD0914 is a novel bacterial gyrase inhibitor that possesses potent in vitro activities against isolates with high-level resistance to ciprofloxacin and extended-spectrum cephalosporins, and it is currently in clinical development for the treatment of N. gonorrhoeae infections. The propensity to develop resistance against AZD0914 was examined in N. gonorrhoeae and found to be extremely low, a finding supported by similar studies with Staphylococcus aureus. The genetic characterization of both first-step and second-step mutants that exhibited decreased susceptibilities to AZD0914 identified substitutions in the conserved GyrB TOPRIM domain, confirming DNA gyrase as the primary target of AZD0914 and providing differentiation from fluoroquinolones. The analysis of available bacterial gyrase and topoisomerase IV structures, including those bound to fluoroquinolone and nonfluoroquinolone inhibitors, has allowed the rationalization of the lack of cross-resistance that AZD0914 shares with fluoroquinolones. Microbiological susceptibility data also indicate that the topoisomerase inhibition mechanisms are subtly different between N. gonorrhoeae and other bacterial species. Taken together, these data support the progression of AZD0914 as a novel treatment option for the oral treatment of N. gonorrhoeae infections.


Subject(s)
Barbiturates/pharmacology , Neisseria gonorrhoeae/drug effects , Spiro Compounds/pharmacology , Topoisomerase II Inhibitors/pharmacology , DNA Gyrase/chemistry , DNA Gyrase/genetics , Drug Resistance, Bacterial , Isoxazoles , Microbial Sensitivity Tests , Morpholines , Mutation , Neisseria gonorrhoeae/genetics , Oxazolidinones
10.
Antimicrob Agents Chemother ; 59(1): 467-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25385112

ABSTRACT

AZD0914 is a new spiropyrimidinetrione bacterial DNA gyrase/topoisomerase inhibitor with potent in vitro antibacterial activity against key Gram-positive (Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pneumoniae, Streptococcus pyogenes, and Streptococcus agalactiae), fastidious Gram-negative (Haemophilus influenzae and Neisseria gonorrhoeae), atypical (Legionella pneumophila), and anaerobic (Clostridium difficile) bacterial species, including isolates with known resistance to fluoroquinolones. AZD0914 works via inhibition of DNA biosynthesis and accumulation of double-strand cleavages; this mechanism of inhibition differs from those of other marketed antibacterial compounds. AZD0914 stabilizes and arrests the cleaved covalent complex of gyrase with double-strand broken DNA under permissive conditions and thus blocks religation of the double-strand cleaved DNA to form fused circular DNA. Whereas this mechanism is similar to that seen with fluoroquinolones, it is mechanistically distinct. AZD0914 exhibited low frequencies of spontaneous resistance in S. aureus, and if mutants were obtained, the mutations mapped to gyrB. Additionally, no cross-resistance was observed for AZD0914 against recent bacterial clinical isolates demonstrating resistance to fluoroquinolones or other drug classes, including macrolides, ß-lactams, glycopeptides, and oxazolidinones. AZD0914 was bactericidal in both minimum bactericidal concentration and in vitro time-kill studies. In in vitro checkerboard/synergy testing with 17 comparator antibacterials, only additivity/indifference was observed. The potent in vitro antibacterial activity (including activity against fluoroquinolone-resistant isolates), low frequency of resistance, lack of cross-resistance, and bactericidal activity of AZD0914 support its continued development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Barbiturates/pharmacology , DNA Gyrase/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Spiro Compounds/pharmacology , Topoisomerase II Inhibitors/pharmacology , Atypical Bacterial Forms/drug effects , Drug Resistance, Bacterial , Fluoroquinolones/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Isoxazoles , Microbial Sensitivity Tests , Morpholines , Oxazolidinones
11.
J Med Chem ; 57(21): 9078-95, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25286019

ABSTRACT

The compounds described herein with a spirocyclic architecture fused to a benzisoxazole ring represent a new class of antibacterial agents that operate by inhibition of DNA gyrase as corroborated in an enzyme assay and by the inhibition of precursor thymidine into DNA during cell growth. Activity resided in the configurationally lowest energy (2S,4R,4aR) diastereomer. Highly active compounds against Staphylococcus aureus had sufficiently high solubility, high plasma protein free fraction, and favorable pharmacokinetics to suggest that in vivo efficacy could be demonstrated, which was realized with compound (-)-1 in S. aureus mouse infection models. A high drug exposure NOEL on oral dosing in the rat suggested that a high therapeutic margin could be achieved. Importantly, (-)-1 was not cross-resistant with other DNA gyrase inhibitors such as fluoroquinolone and aminocoumarin antibacterials. Hence, this class shows considerable promise for the treatment of infections caused by multidrug resistant bacteria, including S. aureus.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Barbiturates/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Isoxazoles/chemical synthesis , Topoisomerase II Inhibitors/chemical synthesis , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Barbiturates/pharmacokinetics , Barbiturates/therapeutic use , Female , Fluoroquinolones/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Inhibitory Concentration 50 , Isoxazoles/pharmacokinetics , Isoxazoles/therapeutic use , Male , Mice , Pyridones/chemical synthesis , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Rats, Wistar , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacokinetics , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Stereoisomerism , Structure-Activity Relationship , Topoisomerase II Inhibitors/pharmacokinetics , Topoisomerase II Inhibitors/therapeutic use
12.
Antimicrob Agents Chemother ; 57(12): 6005-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24041904

ABSTRACT

Inhibitors of 4'-phosphopantetheine adenylyltransferase (PPAT) were identified through high-throughput screening of the AstraZeneca compound library. One series, cycloalkyl pyrimidines, showed inhibition of PPAT isozymes from several species, with the most potent inhibition of enzymes from Gram-positive species. Mode-of-inhibition studies with Streptococcus pneumoniae and Staphylococcus aureus PPAT demonstrated representatives of this series to be reversible inhibitors competitive with phosphopantetheine and uncompetitive with ATP, binding to the enzyme-ATP complex. The potency of this series was optimized using structure-based design, and inhibition of cell growth of Gram-positive species was achieved. Mode-of-action studies, using generation of resistant mutants with targeted sequencing as well as constructs that overexpress PPAT, demonstrated that growth suppression was due to inhibition of PPAT. An effect on bacterial burden was demonstrated in mouse lung and thigh infection models, but further optimization of dosing requirements and compound properties is needed before these compounds can be considered for progress into clinical development. These studies validated PPAT as a novel target for antibacterial therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Animals , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding, Competitive , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemistry , Female , Lung/drug effects , Lung/microbiology , Mice , Models, Molecular , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Pantetheine/analogs & derivatives , Pantetheine/chemistry , Pneumococcal Infections/drug therapy , Pneumococcal Infections/microbiology , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/microbiology , Small Molecule Libraries/chemistry , Staphylococcus aureus/enzymology , Staphylococcus aureus/growth & development , Streptococcus pneumoniae/enzymology , Streptococcus pneumoniae/growth & development , Thigh/microbiology
13.
Bioorg Med Chem Lett ; 23(8): 2362-7, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23499237

ABSTRACT

Lipopolysaccharide (LPS) biosynthesis is an attractive antibacterial target as it is both conserved and essential for the survival of key pathogenic bacteria. Lipid A is the hydrophobic anchor for LPS and a key structural component of the outer membrane of Gram-negative bacteria. Lipid A biosynthesis is performed in part by a unique zinc dependent metalloamidase, LpxC (UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase), which catalyzes the first non-reversible step in lipid A biosynthesis. The UDP portion of the LpxC substrate-binding pocket has been relatively unexplored. We have designed and evaluated a series of hydroxamate based inhibitors which explore the SAR of substitutions directed into the UDP pocket with a range of substituted α-amino acid based linkers. We also provide the first wild type structure of Pseudomonas aeruginosa LpxC which was utilized in the design of many of these analogs.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/chemistry , Amino Acids, Cyclic/chemistry , Uridine Diphosphate/chemistry , Amidohydrolases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Binding Sites , Drug Design , Hydrophobic and Hydrophilic Interactions , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Lipid A/antagonists & inhibitors , Lipid A/biosynthesis , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/biosynthesis , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Uridine Diphosphate/metabolism
14.
ACS Chem Biol ; 7(11): 1866-72, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-22908966

ABSTRACT

There is an urgent need for new antibacterials that pinpoint novel targets and thereby avoid existing resistance mechanisms. We have created novel synthetic antibacterials through structure-based drug design that specifically target bacterial thymidylate kinase (TMK), a nucleotide kinase essential in the DNA synthesis pathway. A high-resolution structure shows compound TK-666 binding partly in the thymidine monophosphate substrate site, but also forming new induced-fit interactions that give picomolar affinity. TK-666 has potent, broad-spectrum Gram-positive microbiological activity (including activity against methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus), bactericidal action with rapid killing kinetics, excellent target selectivity over the human ortholog, and low resistance rates. We demonstrate in vivo efficacy against S. aureus in a murine infected-thigh model. This work presents the first validation of TMK as a compelling antibacterial target and provides a rationale for pursuing novel clinical candidates for treating Gram-positive infections through TMK.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/enzymology , Nucleoside-Phosphate Kinase/antagonists & inhibitors , Enterococcus/drug effects , Enterococcus/enzymology , Gram-Positive Bacterial Infections/drug therapy , Humans , Models, Molecular , Nucleoside-Phosphate Kinase/metabolism , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology
15.
Antimicrob Agents Chemother ; 49(7): 3009-10, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15980386

ABSTRACT

Susceptibility testing with a variety of structurally unrelated compounds showed that hefC in Helicobacter pylori is involved in multidrug efflux. This efflux was shown to depend on the proton motive force, as demonstrated by ethidium bromide accumulation experiments. Thus, H. pylori contains an active multidrug efflux mechanism.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Drug Resistance, Multiple, Bacterial , Helicobacter pylori/drug effects , Proton-Motive Force , Bacterial Outer Membrane Proteins/metabolism , Biological Transport , Ethidium/metabolism , Helicobacter pylori/genetics , Helicobacter pylori/metabolism , Humans , Microbial Sensitivity Tests/methods
SELECTION OF CITATIONS
SEARCH DETAIL