Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Med Chem ; 61(17): 7814-7826, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30086230

ABSTRACT

The spread of antibiotic-resistant pathogens is a growing concern, and new families of antibacterials are desperately needed. Odilorhabdins are a new class of antibacterial compounds that bind to the bacterial ribosome and kill bacteria through inhibition of the translation. NOSO-95C, one of the first member of this family, was synthesized for the first time, and then a structure-activity relationships study was performed to understand which groups are important for antibacterial activity and for inhibition of the bacterial translation. Based on this study an analogue showing improved properties compared to the parent compound was identified and showed promising in vitro and in vivo efficacy against Enterobacteriaceae.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/drug effects , Respiratory Tract Infections/drug therapy , Ribosome Subunits, Small/drug effects , Animals , Humans , Klebsiella Infections/complications , Klebsiella Infections/microbiology , Mice , Microbial Sensitivity Tests , Molecular Structure , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Biosynthesis/drug effects , Respiratory Tract Infections/microbiology , Structure-Activity Relationship , Xenorhabdus
2.
Article in English | MEDLINE | ID: mdl-29987155

ABSTRACT

Antibacterial activity screening of a collection of Xenorhabdus strains led to the discovery of the odilorhabdins, a new antibiotic class with broad-spectrum activity against Gram-positive and Gram-negative pathogens. Odilorhabdins inhibit bacterial translation by a new mechanism of action on ribosomes. A lead optimization program identified NOSO-502 as a promising candidate. NOSO-502 has MIC values ranging from 0.5 to 4 µg/ml against standard Enterobacteriaceae strains and carbapenem-resistant Enterobacteriaceae (CRE) isolates that produce KPC, AmpC, or OXA enzymes and metallo-ß-lactamases. In addition, this compound overcomes multiple chromosome-encoded or plasmid-mediated resistance mechanisms of acquired resistance to colistin. It is effective in mouse systemic infection models against Escherichia coli EN122 (extended-spectrum ß-lactamase [ESBL]) or E. coli ATCC BAA-2469 (NDM-1), achieving a 50% effective dose (ED50) of 3.5 mg/kg of body weight and 1-, 2-, and 3-log reductions in blood burden at 2.6, 3.8, and 5.9 mg/kg, respectively, in the first model and 100% survival in the second, starting with a dose as low as 4 mg/kg. In a urinary tract infection (UTI) model with E. coli UTI89, urine, bladder, and kidney burdens were reduced by 2.39, 1.96, and 1.36 log10 CFU/ml, respectively, after injection of 24 mg/kg. There was no cytotoxicity against HepG2, HK-2, or human renal proximal tubular epithelial cells (HRPTEpiC), no inhibition of hERG-CHO or Nav 1.5-HEK current, and no increase of micronuclei at 512 µM. NOSO-502, a compound with a new mechanism of action, is active against Enterobacteriaceae, including all classes of CRE, has a low potential for resistance development, shows efficacy in several mouse models, and has a favorable in vitro safety profile.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbapenem-Resistant Enterobacteriaceae/drug effects , Escherichia coli/drug effects , Animals , Bacterial Proteins/metabolism , CHO Cells , Carbapenem-Resistant Enterobacteriaceae/metabolism , Cell Line , Cell Line, Tumor , Colistin/pharmacology , Cricetulus , Dogs , Enterobacteriaceae Infections/drug therapy , Escherichia coli/metabolism , Haplorhini , Hep G2 Cells , Humans , Mice , Microbial Sensitivity Tests/methods , Plasmids/metabolism , Rats , beta-Lactamases/metabolism
3.
Mol Cell ; 70(1): 83-94.e7, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29625040

ABSTRACT

Growing resistance of pathogenic bacteria and shortage of antibiotic discovery platforms challenge the use of antibiotics in the clinic. This threat calls for exploration of unconventional sources of antibiotics and identification of inhibitors able to eradicate resistant bacteria. Here we describe a different class of antibiotics, odilorhabdins (ODLs), produced by the enzymes of the non-ribosomal peptide synthetase gene cluster of the nematode-symbiotic bacterium Xenorhabdus nematophila. ODLs show activity against Gram-positive and Gram-negative pathogens, including carbapenem-resistant Enterobacteriaceae, and can eradicate infections in animal models. We demonstrate that the bactericidal ODLs interfere with protein synthesis. Genetic and structural analyses reveal that ODLs bind to the small ribosomal subunit at a site not exploited by current antibiotics. ODLs induce miscoding and promote hungry codon readthrough, amino acid misincorporation, and premature stop codon bypass. We propose that ODLs' miscoding activity reflects their ability to increase the affinity of non-cognate aminoacyl-tRNAs to the ribosome.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacterial Proteins/biosynthesis , DNA, Bacterial/genetics , Klebsiella Infections/drug therapy , Ribosome Subunits, Small/drug effects , Xenorhabdus/metabolism , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , Anti-Bacterial Agents/metabolism , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , Binding Sites , Disease Models, Animal , Female , Hep G2 Cells , Humans , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Male , Mice, Inbred ICR , Protein Biosynthesis/drug effects , Ribosome Subunits, Small/genetics , Ribosome Subunits, Small/metabolism
4.
J Antibiot (Tokyo) ; 66(10): 617-20, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23756685

ABSTRACT

Since the early 1980s, fungi have emerged as a major cause of human disease. Fungal infections are associated with high levels of morbidity and mortality, and are now recognized as an important public health problem. Gram-negative bacterial strains of genus Xenorhabdus are known to form symbiotic associations with soil-dwelling nematodes of the Steinernematidae family. We describe here the discovery of a new antifungal metabolite, cabanillasin, produced by Xenorhabdus cabanillasii. We purified this molecule by cation-exchange chromatography and reverse-phase chromatography. We then determined the chemical structure of cabanillasin by homo- and heteronuclear NMR and MS-MS. Cabanillasin was found to be active against yeasts and filamentous fungi involved in opportunistic infections.


Subject(s)
Antifungal Agents , Fungi/drug effects , Mycoses/microbiology , Opportunistic Infections/microbiology , Xenorhabdus/classification , Xenorhabdus/metabolism , Animals , Antifungal Agents/chemistry , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Cell Line/drug effects , Cross Infection/microbiology , Fungi/classification , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Microbial Sensitivity Tests , Nematoda/microbiology , Xenorhabdus/growth & development
5.
Curr Med Chem ; 16(3): 390-3, 2009.
Article in English | MEDLINE | ID: mdl-19149585

ABSTRACT

Ensuring the availability of new antibiotics to eradicate resistant pathogens is a critical issue, but very few new antibacterials have been recently commercialized. In an effort to rationalize their discovery process, the industry has utilized chemical library and high-throughput approaches already applied in other therapeutical areas to generate new antibiotics. This strategy has turned out to be poorly adapted to the reality of antibacterial discovery. Commercial chemical libraries contain molecules with specific molecular properties, and unfortunately systemic antibacterials are more hydrophilic and have more complex structures. These factors are critical, since hydrophobic antibiotics are generally inactive in the presence of serum. Here, we review how the skewed distribution of systemic antibiotics in chemical space influences the discovery process.


Subject(s)
Anti-Bacterial Agents , Drug Design , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Binding Sites , Biological Products/chemistry , Combinatorial Chemistry Techniques , Humans , Protein Binding , Serum Albumin/chemistry , Serum Albumin/metabolism
6.
Antimicrob Agents Chemother ; 53(4): 1528-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19164148

ABSTRACT

The pharmacologic effect of an antibiotic is directly related to its unbound concentration at the site of infection. Most commercial antibiotics have been selected in part for their low propensity to interact with serum proteins. These nonspecific interactions are classically evaluated by measuring the MIC in the presence of serum. As higher-throughput technologies tend to lose information, surface plasmon resonance (SPR) is emerging as an informative medium-throughput technology for hit validation. Here we show that SPR is a useful automatic tool for quantification of the interaction of model antibiotics with serum proteins and that it delivers precise real-time kinetic data on this critical parameter.


Subject(s)
Anti-Bacterial Agents/metabolism , Blood Proteins/metabolism , Surface Plasmon Resonance/methods , Protein Binding
7.
J Med Chem ; 50(17): 4195-204, 2007 Aug 23.
Article in English | MEDLINE | ID: mdl-17665895

ABSTRACT

The dramatic rise of antibiotic-resistant bacteria over the past two decades has stressed the need for completely novel classes of antibacterial agents. Accordingly, recent advances in the study of prokaryotic transcription open new opportunities for such molecules. This paper reports the structure-activity relationships of a series of phenyl-furanyl-rhodanines (PFRs) as antibacterial inhibitors of RNA polymerase (RNAP). The molecules have been evaluated for their ability to inhibit transcription and affect growth of bacteria living in suspension or in a biofilm and for their propensity to interact with serum albumin, a critical parameter for antibacterial drug discovery. The most active of these molecules inhibit Escherichia coli RNAP transcription at concentrations of

Subject(s)
Anti-Bacterial Agents/chemical synthesis , Biofilms , DNA-Directed RNA Polymerases/antagonists & inhibitors , Furans/chemical synthesis , Gram-Positive Bacteria/drug effects , Rhodanine/analogs & derivatives , Rhodanine/chemical synthesis , Animals , Anti-Bacterial Agents/pharmacology , CHO Cells , Cricetinae , Cricetulus , Furans/pharmacology , Gram-Positive Bacteria/isolation & purification , Gram-Positive Bacteria/physiology , Microbial Sensitivity Tests , Rhodanine/pharmacology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/isolation & purification , Staphylococcus epidermidis/physiology , Structure-Activity Relationship
8.
Antimicrob Agents Chemother ; 51(9): 3117-21, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606690

ABSTRACT

Staphylococcus epidermidis is a major cause of nosocomial infections because of its ability to form biofilms on the surface of medical devices. Only a few antibacterial agents are relatively active against biofilms, and rifampin, a transcription inhibitor, ranks among the most effective molecules against biofilm-related infections. Whether this efficacy is due to advantageous structural properties of rifampin or to the fact that the RNA polymerase is a favorable target remains unclear. In an attempt to answer this question, we investigated the action of different transcription inhibitors against S. epidermidis biofilm, including the newest synthetic transcription inhibitors. This comparison suggests that most of the antibiotics that target the RNA polymerase are active on S. epidermidis biofilms at concentrations close to their MICs. One of these compounds, CBR703, despite its high MIC ranks among the best antibiotics to eradicate biofilm-embedded bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/physiology , Transcription, Genetic/drug effects , Amidines/pharmacology , Colony Count, Microbial , DNA-Directed RNA Polymerases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hydroxylamines/pharmacology , Luminescence , Rifampin/analogs & derivatives , Rifampin/pharmacology , Structure-Activity Relationship
9.
Drug Discov Today ; 12(5-6): 200-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17331884

ABSTRACT

The bacterial RNA polymerase (RNAP) is an essential enzyme that is responsible for making RNA from a DNA template and is targeted by several antibiotics. Rifampicin was the first of such antibiotics to be described and is one of the most efficient anti-tuberculosis drugs in use. In the past five years, structural studies of bacterial RNAP and the resolution of several complexes of drugs bound to RNAP subunits have revealed molecular details of the drug-binding sites and the mechanism of drug action. This knowledge opens avenues for the development of antibiotics. Here these drugs are reviewed, together with their mechanisms and their potential interest for therapeutic applications.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Delivery Systems , Drug Design , Transcription, Genetic/drug effects , Antibiotics, Antitubercular/pharmacology , Binding Sites/genetics , DNA-Directed RNA Polymerases , Humans , Protein Subunits/genetics , RNA, Bacterial/drug effects , Rifampin/pharmacology , Structure-Activity Relationship
10.
J Antimicrob Chemother ; 58(4): 778-83, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16880176

ABSTRACT

OBJECTIVES: Staphylococcus epidermidis biofilms form at the surface of implants and prostheses and are responsible for the failure of many antibiotic therapies. Only a few antibiotics are relatively active against biofilms, and rifampicin, a transcription inhibitor, is among the most effective molecules for treating biofilm-related infections. Having recently selected a new potential transcription inhibitor, we attempted to evaluate its efficacy against S. epidermidis biofilms. METHODS: Biofilm-forming S. epidermidis strains were grown planktonically or as biofilms and their susceptibility to this transcription inhibitor was compared with reference antibiotics with different mechanisms of action. CONCLUSIONS: Our results demonstrate that this new molecule is active; its effects are fast and kinetically related to those of rifampicin, but unlike rifampicin it does not select for resistant bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , DNA-Directed RNA Polymerases/antagonists & inhibitors , Rhodanine/analogs & derivatives , Rhodanine/pharmacology , Staphylococcus epidermidis/drug effects , Anti-Bacterial Agents/chemistry , Biofilms/growth & development , Humans , Microbial Sensitivity Tests , Polystyrenes , Rifampin/pharmacology , Staphylococcus epidermidis/growth & development , Transcription, Genetic/drug effects
11.
J Antimicrob Chemother ; 57(2): 245-51, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16373430

ABSTRACT

OBJECTIVES: Despite extensive functional screening of the bacterial RNA polymerase (RNAP) over the past years, very few novel inhibitors have been reported. We have, therefore, decided to screen with a radically different, non-enzymic, protein-protein interaction assay. Our target is the highly conserved RNAP-sigma interaction that is essential for transcription. METHODS: Small molecule inhibitors of the RNAP-sigma interaction were tested for their activity on transcription and on bacteria. RESULTS: These compounds have antibacterial activity against Gram-positive bacteria including multiresistant clinical isolates. CONCLUSIONS: This is, to our knowledge, the first example of a small molecule inhibitor of this interaction.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/enzymology , DNA-Directed RNA Polymerases/drug effects , Bacillus anthracis/drug effects , Bacillus cereus/drug effects , Drug Resistance, Bacterial , Enzyme-Linked Immunosorbent Assay , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Immunoprecipitation , Microbial Sensitivity Tests , Staphylococcus epidermidis/drug effects , Streptococcus pneumoniae/drug effects , Transcription, Genetic/drug effects
13.
Assay Drug Dev Technol ; 2(6): 629-35, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15674021

ABSTRACT

We have developed a multiwell assay for the detection of modulators of prokaryotic transcription based on the quantification of protein-protein interaction. This assay consists of three steps: (a) the immobilization of the Escherichia coli protein sigma70 in the well, (b) the incubation of the immobilized protein with core RNA polymerase and a potential inhibitor, and (c) washing and quantification of the binding of core to sigma70 with a monoclonal antibody conjugated to horseradish peroxidase. We show that this assay is sensitive, reproducible, and robust, and is able to discriminate between control competitors with different affinities. We demonstrate the usefulness of the assay to screen for microbial RNA polymerase inhibitors as potential new drugs for the treatment of emerging antibiotic-resistant bacteria.


Subject(s)
DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/biosynthesis , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Escherichia coli/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Chromatography, Gel , DNA-Directed RNA Polymerases/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Gene Library , Horseradish Peroxidase/chemistry , Horseradish Peroxidase/pharmacology , Reproducibility of Results , Sigma Factor/metabolism , Solvents , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...