Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Antimicrob Agents Chemother ; 59(3): 1455-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534737

RESUMEN

New drugs to treat drug-resistant tuberculosis are urgently needed. Extensively drug-resistant and probably the totally drug-resistant tuberculosis strains are resistant to fluoroquinolones like moxifloxacin, which target gyrase A, and most people infected with these strains die within a year. In this study, we found that a novel aminobenzimidazole, VXc-486, which targets gyrase B, potently inhibits multiple drug-sensitive isolates and drug-resistant isolates of Mycobacterium tuberculosis in vitro (MICs of 0.03 to 0.30 µg/ml and 0.08 to 5.48 µg/ml, respectively) and reduces mycobacterial burdens in lungs of infected mice in vivo. VXc-486 is active against drug-resistant isolates, has bactericidal activity, and kills intracellular and dormant M. tuberculosis bacteria in a low-oxygen environment. Furthermore, we found that VXc-486 inhibits the growth of multiple strains of Mycobacterium abscessus, Mycobacterium avium complex, and Mycobacterium kansasii (MICs of 0.1 to 2.0 µg/ml), as well as that of several strains of Nocardia spp. (MICs of 0.1 to 1.0 µg/ml). We made a direct comparison of the parent compound VXc-486 and a phosphate prodrug of VXc-486 and showed that the prodrug of VXc-486 had more potent killing of M. tuberculosis than did VXc-486 in vivo. In combination with other antimycobacterial drugs, the prodrug of VXc-486 sterilized M. tuberculosis infection when combined with rifapentine-pyrazinamide and bedaquiline-pyrazinamide in a relapse infection study in mice. Furthermore, the prodrug of VXc-486 appeared to perform at least as well as the gyrase A inhibitor moxifloxacin. These findings warrant further development of the prodrug of VXc-486 for the treatment of tuberculosis and nontuberculosis mycobacterial infections.


Asunto(s)
Antibacterianos/uso terapéutico , Bencimidazoles/uso terapéutico , Infecciones por Mycobacterium/tratamiento farmacológico , Inhibidores de Topoisomerasa II/uso terapéutico , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana
2.
J Antimicrob Chemother ; 69(7): 1866-72, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24651828

RESUMEN

OBJECTIVES: Clinical resistance to the currently recommended extended-spectrum cephalosporins (ESCs), the last remaining options for empirical antimicrobial monotherapy of gonorrhoea globally, has been reported. New antimicrobials are essential to avoid the emergence of untreatable gonorrhoea. We have investigated the in vitro activity of a novel dual bacterial topoisomerase inhibitor of the ATPase activities of GyrB and ParE (Vertex aminobenzimidazole VT12-008911), compared with antimicrobials currently or previously recommended for gonorrhoea treatment. METHODS: MICs were determined using agar dilution (VT12-008911) or Etest (seven antimicrobials) for international reference strains (n = 28) and clinical Neisseria gonorrhoeae isolates (n = 220). The latter included three extensively drug-resistant isolates with high-level ceftriaxone resistance, additional isolates with clinical ESC resistance and a high number of isolates with ciprofloxacin resistance and multidrug resistance. RESULTS: The MIC(50), MIC(90) and MIC range of VT12-008911 were 0.064, 0.125 and ≤0.002-0.25 mg/L, respectively. One-hundred and seventy (69%) isolates were ciprofloxacin resistant; however, only 54 of those isolates had a VT12-008911 MIC >0.064 mg/L (47 and 7 with MIC = 0.125 mg/L and MIC = 0.25 mg/L, respectively). The in vitro activity of VT12-008911 was superior to that of ciprofloxacin and all additional antimicrobials investigated. Time-kill curve analysis showed that VT12-008911 exhibited potent time-dependent bactericidal activity, at or very close to the MIC, against N. gonorrhoeae. CONCLUSIONS: In vitro results suggest that VT12-008911 might be an effective treatment option for gonorrhoea. However, it will be important to detail the pharmacokinetics/pharmacodynamics, toxicity, selection and mechanisms of VT12-008911 resistance in N. gonorrhoeae and, finally, to perform well-designed in vivo randomized clinical trials.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Antibacterianos/farmacología , Girasa de ADN/metabolismo , Topoisomerasa de ADN IV/antagonistas & inhibidores , Farmacorresistencia Bacteriana , Neisseria gonorrhoeae/efectos de los fármacos , Inhibidores de Topoisomerasa/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Neisseria gonorrhoeae/enzimología
3.
Antimicrob Agents Chemother ; 57(12): 6236-45, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24100495

RESUMEN

Telaprevir is a linear, peptidomimetic small molecule that inhibits hepatitis C virus (HCV) replication by specifically inhibiting the NS3·4A protease. In phase 3 clinical studies, telaprevir in combination with peginterferon and ribavirin (PR) significantly improved sustained virologic response (SVR) rates in genotype 1 chronic HCV-infected patients compared with PR alone. In patients who do not achieve SVR after treatment with telaprevir-based regimens, variants with mutations in the NS3·4A protease region have been observed. Such variants can contribute to drug resistance and limit the efficacy of treatment. To gain a better understanding of the viral resistance profile, we conducted phenotypic characterization of the variants using HCV replicons carrying site-directed mutations. The most frequently observed (significantly enriched) telaprevir-resistant variants, V36A/M, T54A/S, R155K/T, and A156S, conferred lower-level resistance (3- to 25-fold), whereas A156T and V36M+R155K conferred higher-level resistance (>25-fold) to telaprevir. Rarely observed (not significantly enriched) variants included V36I/L and I132V, which did not confer resistance to telaprevir; V36C/G, R155G/I/M/S, V36A+T54A, V36L+R155K, T54S+R155K, and R155T+D168N, which conferred lower-level resistance to telaprevir; and A156F/N/V, V36A+R155K/T, V36M+R155T, V36A/M+A156T, T54A+A156S, T54S+A156S/T, and V36M+T54S+R155K, which conferred higher-level resistance to telaprevir. All telaprevir-resistant variants remained fully sensitive to alpha interferon, ribavirin, and HCV NS5B nucleoside and nonnucleoside polymerase inhibitors. In general, the replication capacity of telaprevir-resistant variants was lower than that of the wild-type replicon.


Asunto(s)
Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Oligopéptidos/farmacología , Proteínas no Estructurales Virales/genética , Antivirales/farmacología , Línea Celular , Hepacivirus/genética , Humanos , Concentración 50 Inhibidora , Mutagénesis Sitio-Dirigida , Inhibidores de Proteasas/farmacología
4.
ACS Med Chem Lett ; 8(12): 1224-1229, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29259738

RESUMEN

Drug resistant tuberculosis (TB) infections are on the rise and antibiotics that inhibit Mycobacterium tuberculosis through a novel mechanism could be an important component of evolving TB therapy. Protein kinase A (PknA) and protein kinase B (PknB) are both essential serine-threonine kinases in M. tuberculosis. Given the extensive knowledge base in kinase inhibition, these enzymes present an interesting opportunity for antimycobacterial drug discovery. This study focused on targeting both PknA and PknB while improving the selectivity window over related mammalian kinases. Compounds achieved potent inhibition (Ki ≈ 5 nM) of both PknA and PknB. A binding pocket unique to mycobacterial kinases was identified. Substitutions that filled this pocket resulted in a 100-fold differential against a broad selection of mammalian kinases. Reducing lipophilicity improved antimycobacterial activity with the most potent compounds achieving minimum inhibitory concentrations ranging from 3 to 5 µM (1-2 µg/mL) against the H37Ra isolate of M. tuberculosis.

5.
ACS Med Chem Lett ; 6(7): 822-6, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26191374

RESUMEN

Benzimidazole 1 is the lead compound resulting from an antibacterial program targeting dual inhibitors of bacterial DNA gyrase and topoisomerase IV. With the goal of improving key drug-like properties, namely, the solubility and the formulability of 1, an effort to identify prodrugs was undertaken. This has led to the discovery of a phosphate ester prodrug 2. This prodrug is rapidly cleaved to the parent drug molecule upon both oral and intravenous administration. The prodrug achieved equivalent exposure of 1 compared to dosing the parent in multiple species. The prodrug 2 has improved aqueous solubility, simplifying both intravenous and oral formulation.

6.
J Med Chem ; 57(21): 8792-816, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25317480

RESUMEN

Compound 3 is a potent aminobenzimidazole urea with broad-spectrum Gram-positive antibacterial activity resulting from dual inhibition of bacterial gyrase (GyrB) and topoisomerase IV (ParE), and it demonstrates efficacy in rodent models of bacterial infection. Preclinical in vitro and in vivo studies showed that compound 3 covalently labels liver proteins, presumably via formation of a reactive metabolite, and hence presented a potential safety liability. The urea moiety in compound 3 was identified as being potentially responsible for reactive metabolite formation, but its replacement resulted in loss of antibacterial activity and/or oral exposure due to poor physicochemical parameters. To identify second-generation aminobenzimidazole ureas devoid of reactive metabolite formation potential, we implemented a metabolic shift strategy, which focused on shifting metabolism away from the urea moiety by introducing metabolic soft spots elsewhere in the molecule. Aminobenzimidazole urea 34, identified through this strategy, exhibits similar antibacterial activity as that of 3 and did not label liver proteins in vivo, indicating reduced/no potential for reactive metabolite formation.


Asunto(s)
Antibacterianos/síntesis química , Bencimidazoles/síntesis química , Inhibidores Enzimáticos/síntesis química , Animales , Antibacterianos/metabolismo , Bencimidazoles/metabolismo , Girasa de ADN/metabolismo , Topoisomerasa de ADN IV/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores Enzimáticos/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Microsomas Hepáticos/metabolismo , Relación Estructura-Actividad , Inhibidores de Topoisomerasa II/metabolismo , Urea/análogos & derivados , Urea/síntesis química , Urea/metabolismo
7.
Antimicrob Agents Chemother ; 52(1): 110-20, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17938182

RESUMEN

In patients chronically infected with hepatitis C virus (HCV) strains of genotype 1, rapid and dramatic antiviral activity has been observed with telaprevir (VX-950), a highly selective and potent inhibitor of the HCV NS3-4A serine protease. HCV variants with substitutions in the NS3 protease domain were observed in some patients during telaprevir dosing. In this study, purified protease domain proteins and reconstituted HCV subgenomic replicons were used for phenotypic characterization of many of these substitutions. V36A/M or T54A substitutions conferred less than eightfold resistance to telaprevir. Variants with double substitutions at Val36 plus Thr54 had approximately 20-fold resistance to telaprevir, and variants with double substitutions at Val36 plus Arg155 or Ala156 had >40-fold resistance to telaprevir. An X-ray structure of the HCV strain H protease domain containing the V36M substitution in a cocomplex with an NS4A cofactor peptide was solved at a 2.4-A resolution. Except for the side chain of Met36, the V36M variant structure is identical to that of the wild-type apoenzyme. The in vitro replication capacity of most variants was significantly lower than that of the wild-type replicon in cells, which is consistent with the impaired in vivo fitness estimated from telaprevir-dosed patients. Finally, the sensitivity of these replicon variants to alpha interferon or ribavirin remained unchanged compared to that of the wild-type.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/genética , Variación Genética , Hepacivirus/efectos de los fármacos , Oligopéptidos/farmacología , Proteínas no Estructurales Virales , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Antivirales/uso terapéutico , Línea Celular , Cristalografía por Rayos X , Hepacivirus/clasificación , Hepacivirus/enzimología , Hepacivirus/fisiología , Hepatitis C/tratamiento farmacológico , Hepatitis C/virología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Oligopéptidos/uso terapéutico , Fenotipo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
8.
J Biol Chem ; 282(31): 22619-28, 2007 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-17556358

RESUMEN

Telaprevir (VX-950) is a highly selective, potent inhibitor of the hepatitis C virus (HCV) NS3.4A serine protease. It has demonstrated strong antiviral activity in patients chronically infected with genotype 1 HCV when dosed alone or in combination with peginterferon alfa-2a. Substitutions of Arg(155) of the HCV NS3 protease domain have been previously detected in HCV isolates from some patients during telaprevir dosing. In this study, Arg(155) was replaced with various residues in genotype 1a protease domain proteins and in genotype 1b HCV subgenomic replicons. Characterization of both the purified enzymes and reconstituted replicon cells demonstrated that substitutions of Arg(155) with these residues conferred low level resistance to telaprevir (<25-fold). An x-ray structure of genotype 1a HCV protease domain with the R155K mutation, in a complex with an NS4A co-factor peptide, was determined at a resolution of 2.5A. The crystal structure of the R155K protease is essentially identical to that of the wild-type apoenzyme (Protein Data Bank code 1A1R) except for the side chain of mutated residue 155. Telaprevir was docked into the x-ray structure of the R155K protease, and modeling analysis suggests that the P2 group of telaprevir loses several hydrophobic contacts with the Lys(155) side chain. It was demonstrated that replicon cells containing substitutions at NS3 protease residue 155 remain fully sensitive to interferon alpha or ribavirin. Finally, these variant replicons were shown to have reduced replication capacity compared with the wild-type HCV replicon in cells.


Asunto(s)
Antivirales/química , Arginina/química , Interferón-alfa/química , Oligopéptidos/química , Polietilenglicoles/química , Ribavirina/química , Proteínas no Estructurales Virales/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Concentración 50 Inhibidora , Interferón alfa-2 , Modelos Moleculares , Datos de Secuencia Molecular , Fenotipo , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes
9.
Gastroenterology ; 132(5): 1767-77, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17484874

RESUMEN

BACKGROUND & AIMS: Telaprevir (VX-950), a hepatitis C virus (HCV) NS3.4A protease inhibitor, has shown strong antiviral activity in phase 1 clinical studies. Because of high levels of HCV replication and the low fidelity of HCV polymerase, selection of resistant isolates during therapy may occur. METHODS: A highly sensitive sequencing method was developed in which approximately 80 clones/sample were analyzed to identify mutations in the NS3 protease catalytic domain in HCV genotype-1-infected patients dosed with 450 mg every 8 hours, 750 mg every 8 hours, or 1250 mg every 12 hours of telaprevir for 14 days. RESULTS: Mutations that confer low-level resistance (V36A/M, T54A, R155K/T, and A156S) and high-level resistance (A156V/T, 36+155, 36+156) to telaprevir were detected and correlated with telaprevir exposure and virologic response. Changes in the frequency of mutations after the end of dosing showed an inverse relationship between in vivo viral fitness and resistance. In the absence of telaprevir selective pressure the majority of resistant variants were replaced by wild-type virus within 3-7 months. CONCLUSIONS: Resistant HCV isolates are selected rapidly during therapy with the highly active protease inhibitor telaprevir. Combination therapy with pegylated interferon-alfa or other direct antiviral drugs seem mandatory to avoid developing resistance.


Asunto(s)
Genotipo , Hepacivirus/crecimiento & desarrollo , Hepacivirus/genética , Hepatitis C/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Fenotipo , Inhibidores de Proteasas/uso terapéutico , Dominio Catalítico/genética , ADN Viral/genética , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Quimioterapia Combinada , Humanos , Interferón alfa-2 , Interferón-alfa/uso terapéutico , Mutación/genética , Oligopéptidos/farmacología , Polietilenglicoles/uso terapéutico , Inhibidores de Proteasas/farmacología , Proteínas Recombinantes , Serina Endopeptidasas/genética , Factores de Tiempo , Proteínas no Estructurales Virales , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
10.
Proc Natl Acad Sci U S A ; 103(45): 16948-52, 2006 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17075036

RESUMEN

The pathogenic bacterium Pseudomonas aeruginosa uses acyl-homoserine lactone quorum-sensing signals to coordinate the expression of a battery of virulence genes in a cascade of regulatory events. The quorum-sensing signal that triggers the cascade is N-3-oxo-dodecanoyl homoserine lactone (3OC12-HSL), which interacts with two signal receptor-transcription factors, LasR and QscR. This signal is base labile, and it is degraded by mammalian PON lactonases. We have identified a structurally unrelated triphenyl mimic of 3OC12-HSL that is base-insensitive and PON-resistant. The triphenyl mimic seems to interact specifically with LasR but not with QscR. In silico analysis suggests that the mimic fits into the 3OC12-HSL-binding site of LasR and makes key contacts with LasR. The triphenyl mimic is an excellent scaffold for developing quorum-sensing inhibitors, and its stability and potency make it ideal for biotechnology uses such as heterologous gene expression.


Asunto(s)
4-Butirolactona/análogos & derivados , Homoserina/análogos & derivados , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum/fisiología , 4-Butirolactona/química , 4-Butirolactona/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Homoserina/química , Homoserina/metabolismo , Modelos Moleculares , Imitación Molecular , Estructura Molecular , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Transducción de Señal , Transactivadores/química , Transactivadores/metabolismo
11.
Antimicrob Agents Chemother ; 50(4): 1228-37, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16569833

RESUMEN

Antibiotics with novel mechanisms of action are becoming increasingly important in the battle against bacterial resistance to all currently used classes of antibiotics. Bacterial DNA gyrase and topoisomerase IV (topoIV) are the familiar targets of fluoroquinolone and coumarin antibiotics. Here we present the characterization of two members of a new class of synthetic bacterial topoII ATPase inhibitors: VRT-125853 and VRT-752586. These aminobenzimidazole compounds were potent inhibitors of both DNA gyrase and topoIV and had excellent antibacterial activities against a wide spectrum of problematic pathogens responsible for both nosocomial and community-acquired infections, including staphylococci, streptococci, enterococci, and mycobacteria. Consistent with the novelty of their structures and mechanisms of action, antibacterial potency was unaffected by commonly encountered resistance phenotypes, including fluoroquinolone resistance. In time-kill assays, VRT-125853 and VRT-752586 were bactericidal against Staphylococcus aureus, Streptococcus pneumoniae, Enterococcus faecalis, and Haemophilus influenzae, causing 3-log reductions in viable cells within 24 h. Finally, similar to the fluoroquinolones, relatively low frequencies of spontaneous resistance to VRT-125853 and VRT-752586 were found, a property consistent with their in vitro dual-targeting activities.


Asunto(s)
Antibacterianos/farmacología , Bencimidazoles/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores de Topoisomerasa II , Urea/análogos & derivados , Proteínas Sanguíneas/metabolismo , Girasa de ADN , Farmacorresistencia Bacteriana , Humanos , Pruebas de Sensibilidad Microbiana , Urea/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA