Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-30150466

RESUMO

Inhibition of NS5A has emerged as an attractive strategy to intervene in hepatitis C virus (HCV) replication. Ruzasvir (formerly MK-8408) was developed as a novel NS5A inhibitor to improve upon the potency and barrier to resistance of early compounds. Ruzasvir inhibited HCV RNA replication with 50% effective concentrations (EC50s) of 1 to 4 pM in Huh7 or Huh7.5 cells bearing replicons for HCV genotype 1 (GT1) to GT7. The antiviral activity was modestly (10-fold) reduced in the presence of 40% normal human serum. The picomolar potency in replicon cells extended to sequences of clinical isolates available in public databases that were synthesized and tested as replicons. In GT1a, ruzasvir inhibited common NS5A resistance-associated substitutions (RASs), with the exception of M28G. De novo resistance selection studies identified pathways with certain amino acid substitutions at residues 28, 30, 31, and 93 across genotypes. Substitutions at position 93 were more common in GT1 to -4, while changes at position 31 emerged frequently in GT5 and -6. With the exception of GT4, the reintroduction of selected RASs conferred a ≥100-fold potency reduction in the antiviral activity of ruzasvir. Common RASs from other classes of direct-acting antiviral agents (DAAs) did not confer cross-resistance to ruzasvir. The interaction of ruzasvir with an NS3/4A protease inhibitor (grazoprevir) and an NS5B polymerase prodrug (uprifosbuvir) was additive to synergistic, with no evidence of antagonism or cytotoxicity. The antiviral profile of ruzasvir supported its further evaluation in human trials in combination with grazoprevir and uprifosbuvir.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Pirrolidinas/farmacologia , Tiazóis/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Amidas , Carbamatos , Linhagem Celular Tumoral , Ciclopropanos , Farmacorresistência Viral/efeitos dos fármacos , Quimioterapia Combinada/métodos , Genótipo , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/virologia , Humanos , Inibidores de Proteases/farmacologia , Quinoxalinas/farmacologia , Replicon/efeitos dos fármacos , Sulfonamidas , Uridina/análogos & derivados , Uridina/farmacologia
2.
J Biol Chem ; 292(15): 6202-6212, 2017 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-28228479

RESUMO

Grazoprevir is a potent pan-genotype and macrocyclic inhibitor of hepatitis C virus (HCV) NS3/4A protease and was developed for treating chronic HCV infection. In HCV genotype (GT) 1a, grazoprevir maintains potent activity against a majority of NS3 resistance-associated amino acid substitutions, including the highly prevalent and naturally occurring Q80K polymorphism that impacts simeprevir, another NS3/4A protease inhibitor. The basis for an unexpected difference in the clinical impact of some NS3 substitutions was investigated. Phenotypic analysis of resistance-associated substitutions identified in NS3 from GT1a-infected patients who failed therapy with grazoprevir (in combination with elbasvir, an inhibitor of HCV NS5A protein) showed that positions 56, 156, and 168 in NS3 were most impactful because they diminished protein-inhibitor interactions. Although an amino acid substitution from aspartic acid to alanine at position 168 (D168A) reduced the potency of grazoprevir, its combination with R155K unexpectedly nullified this effect. Molecular dynamics and free-energy surface studies indicated that Asp-168 is important in anchoring Arg-155 for ligand binding but is not critical for Lys-155 because of the inherent flexibility of its side chain. Moreover, modeling studies supported a strong direct cation-heterocycle interaction between the Lys-155 side chain of the double substitution, R155K/D168A, and the lone pair on the quinoxaline in grazoprevir. This unique interaction provides a structural basis for grazoprevir's higher potency than simeprevir, an inhibitor to which the double substitution confers a significant reduction in potency. Our findings are consistent with the detection of R155K/D168A in NS3 from virologic failures treated with simeprevir but not grazoprevir.


Assuntos
Hepacivirus/enzimologia , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto , Quinoxalinas/química , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Amidas , Substituição de Aminoácidos , Carbamatos , Linhagem Celular Tumoral , Ciclopropanos , Hepacivirus/genética , Hepatite C/tratamento farmacológico , Hepatite C/enzimologia , Hepatite C/genética , Humanos , Quinoxalinas/uso terapêutico , Simeprevir/química , Simeprevir/uso terapêutico , Sulfonamidas , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
3.
Antimicrob Agents Chemother ; 57(7): 3250-61, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23629699

RESUMO

While new direct-acting antiviral agents for the treatment of chronic hepatitis C virus (HCV) infection have been approved, there is a continued need for novel antiviral agents that act on new targets and can be used in combination with current therapies to enhance efficacy and to restrict the emergence of drug-resistant viral variants. To this end, we have identified a novel class of small molecules, exemplified by PTC725, that target the nonstructural protein 4B (NS4B). PTC725 inhibited HCV 1b (Con1) replicons with a 50% effective concentration (EC50) of 1.7 nM and an EC90 of 9.6 nM and demonstrated a >1,000-fold selectivity window with respect to cytotoxicity. The compounds were fully active against HCV replicon mutants that are resistant to inhibitors of NS3 protease and NS5B polymerase. Replicons selected for resistance to PTC725 harbored amino acid substitutions F98L/C and V105M in NS4B. Anti-replicon activity of PTC725 was additive to synergistic in combination with alpha interferon or with inhibitors of HCV protease and polymerase. Immunofluorescence microscopy demonstrated that neither the HCV inhibitors nor the F98C substitution altered the subcellular localization of NS4B or NS5A in replicon cells. Oral dosing of PTC725 showed a favorable pharmacokinetic profile with high liver and plasma exposure in mice and rats. Modeling of dosing regimens in humans indicates that a once-per-day or twice-per-day oral dosing regimen is feasible. Overall, the preclinical data support the development of PTC725 for use in the treatment of chronic HCV infection.


Assuntos
Antivirais/metabolismo , Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Indóis/farmacologia , Sulfonamidas/farmacologia , Proteínas não Estruturais Virais/metabolismo , Substituição de Aminoácidos , Animais , Antivirais/farmacocinética , Linhagem Celular Tumoral , Farmacorresistência Viral/genética , Sinergismo Farmacológico , Humanos , Indóis/metabolismo , Indóis/farmacocinética , Interferon-alfa/farmacologia , Masculino , Camundongos , Testes de Sensibilidade Microbiana , Ratos , Ratos Sprague-Dawley , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética , Proteínas não Estruturais Virais/genética , Replicação Viral/efeitos dos fármacos
4.
Antiviral Res ; 84(2): 178-84, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19747948

RESUMO

Boceprevir (SCH 503034) is an orally active novel inhibitor of the hepatitis C virus (HCV) NS3 protease currently in clinical development for the treatment of hepatitis C. In this in vitro study, we demonstrate that combination of boceprevir with a nucleoside analog or a non-nucleoside HCV NS5B polymerase inhibitor was superior to treatment by single agents in inhibiting viral RNA replication in replicon cells. In the presence of boceprevir (at 5xEC(90)), the addition of 2'-C-methyl-adenosine or an indole-N-acetamide targeting the polymerase finger-loop site (at 1xEC(90)) significantly reduced the emergence of resistant replicon colonies. A higher dose (5xEC(90)) of either of the polymerase inhibitors in combination with boceprevir suppressed replicon resistance further to below detectable levels. Sequencing analysis of replicon cells selected by the combination treatment revealed known resistance mutations to the two polymerase inhibitors but no previously reported resistance mutations to boceprevir. Interestingly, a novel mutation (M175L) in the protease domain was identified. The dually resistant replicon cells were monitored for over 30 passages and sensitivity to polymerase inhibitors was found to decrease over time in a manner that correlated with the increasing prevalence of specific resistance mutations. Importantly, these cells remained sensitive to interferon-alpha and different classes of polymerase inhibitors. These findings support the rationale for clinical evaluation of combination treatment of HCV protease and polymerase inhibitors.


Assuntos
Inibidores Enzimáticos/farmacologia , Hepacivirus/efeitos dos fármacos , Mutação , Prolina/análogos & derivados , Inibidores de Proteases/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/genética , Antivirais/farmacologia , Antivirais/uso terapêutico , Farmacorresistência Viral , Quimioterapia Combinada , Hepacivirus/enzimologia , Hepacivirus/genética , Hepacivirus/fisiologia , Humanos , Prolina/química , Prolina/farmacologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Replicon/efeitos dos fármacos , Inoculações Seriadas , Replicação Viral/efeitos dos fármacos
5.
Anal Biochem ; 366(1): 9-17, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17493575

RESUMO

DNA ligase catalyzes the closure of single-strand nicks in double-stranded DNA that arise during replication and recombination. Inhibition of bacterial ligase is expected to cause chromosome degradation and cell death, making it an attractive target for new antibacterials. The prototypical bacterial ligase couples the hydrolysis of NAD(+) to phosphodiester bond formation between an adjacent 3'OH and 5'-terminal phosphate of nicked duplex DNA. The first step is the reversible formation of a ligase-adenylate from the reaction between apoenzyme and NAD(+). Inhibitors that compete with NAD(+) are expected to be bacterial specific because eukaryotic DNA ligases use ATP and differ in the sequence composition of their adenylation domain. We report here a high-throughput assay that measures the adenylation reaction specifically by monitoring ligase-AMP formation via scintillation proximity technologies. Escherichia coli DNA ligase was biotinylated in vivo; after reaction with radiolabeled NAD(+), ligase-[(3)H]AMP could be captured onto the streptavidin-coated surface of the solid scintillant. The method was ideal for high-throughput screening because it required minimal manipulations and generated a robust signal with minimal scatter. Certain adenosine analogs were found to inhibit the adenylation assay and had similar potency of inhibition in a DNA ligation assay.


Assuntos
Técnicas de Química Analítica/métodos , DNA Ligases/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Monofosfato de Adenosina/biossíntese , Trifosfato de Adenosina/metabolismo , Sequência de Bases , Biotina , DNA/genética , DNA/metabolismo , DNA Ligases/antagonistas & inibidores , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Escherichia coli/enzimologia , Cinética , Ligantes , NAD/metabolismo , Reprodutibilidade dos Testes , Contagem de Cintilação , Estreptavidina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA