Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Bioorg Med Chem Lett ; 22(15): 4998-5002, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22765892

RESUMO

A new class of benzoxazole and benzothiazole amide derivatives exhibiting potent CYP3A4 inhibiting properties was identified. Extensive lead optimization was aimed at improving the CYP3A4 inhibitory properties as well as overall ADME profile of these amide derivatives. This led to the identification of thiazol-5-ylmethyl (2S,3R)-4-(2-(ethyl(methyl)amino)-N-isobutylbenzo[d]oxazole-6-carboxamido)-3-hydroxy-1-phenylbutan-2-ylcarbamate (C1) as a lead candidate for this class. This compound together with structurally similar analogues demonstrated excellent 'boosting' properties when tested in dogs. These findings warrant further evaluation of their properties in an effort to identify valuable alternatives to Ritonavir as pharmacokinetic enhancers.


Assuntos
Amidas/química , Benzotiazóis/química , Benzoxazóis/química , Inibidores da Protease de HIV/química , Amidas/síntese química , Amidas/farmacocinética , Animais , Células CACO-2 , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A , Cães , Inibidores da Protease de HIV/síntese química , Inibidores da Protease de HIV/farmacocinética , HIV-1/enzimologia , Meia-Vida , Humanos , Ratos , Relação Estrutura-Atividade
2.
Nucleic Acids Res ; 38(6): 1902-12, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20015971

RESUMO

The host restriction factor Apobec3G is a cytidine deaminase that incorporates into HIV-1 virions and interferes with viral replication. The HIV-1 accessory protein Vif subverts Apobec3G by targeting it for proteasomal degradation. We propose a model in which Apobec3G N-terminal domains symmetrically interact via a head-to-head interface containing residues 122 RLYYFW 127. To validate this model and to characterize the Apobec3G-Apobec3G and the Apobec3G-Vif interactions, the mammalian protein-protein interaction trap two-hybrid technique was used. Mutations in the head-to-head interface abrogate the Apobec3G-Apobec3G interaction. All mutations that inhibit Apobec3G-Apobec3G binding also inhibit the Apobec3G-Vif interaction, indicating that the head-to head interface plays an important role in the interaction with Vif. Only the D128K, P129A and T32Q mutations specifically affect the Apobec3G-Vif association. In our model, D128, P129 and T32 cluster at the edge of the head-to-head interface, possibly forming a Vif binding site composed of two Apobec3G molecules. We propose that Vif either binds at the Apobec3G head-to-head interface or associates with an RNA-stabilized Apobec3G oligomer.


Assuntos
Citidina Desaminase/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Desaminase APOBEC-3G , Sítios de Ligação , Linhagem Celular , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Citosina Desaminase/química , Dimerização , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Domínios e Motivos de Interação entre Proteínas , Homologia Estrutural de Proteína , Técnicas do Sistema de Duplo-Híbrido , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo
3.
J Virol ; 82(21): 10366-74, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18715920

RESUMO

Integration of viral DNA into the host chromosome is an essential step in the life cycle of retroviruses and is facilitated by the viral integrase enzyme. The first generation of integrase inhibitors recently approved or currently in late-stage clinical trials shows great promise for the treatment of human immunodeficiency virus (HIV) infection, but virus is expected to develop resistance to these drugs. Therefore, we used a novel resistance selection protocol to follow the emergence of resistant HIV in the presence of the integrase inhibitor elvitegravir (GS-9137). We find the primary resistance-conferring mutations to be Q148R, E92Q, and T66I and demonstrate that they confer a reduction in susceptibility not only to elvitegravir but also to raltegravir (MK-0518) and other integrase inhibitors. The locations of the mutations are highlighted in the catalytic sites of integrase, and we correlate the mutations with expected drug-protein contacts. In addition, mutations that do not confer reduced susceptibility when present alone (H114Y, L74M, R20K, A128T, E138K, and S230R) are also discussed in relation to their position in the catalytic core domain and their proximity to known structural features of integrase. These data broaden the understanding of antiviral resistance against integrase inhibitors and may give insight facilitating the discovery of second-generation compounds.


Assuntos
Farmacorresistência Viral , Integrase de HIV/genética , HIV-1/efeitos dos fármacos , HIV-1/genética , Inibidores de Integrase/farmacologia , Mutação de Sentido Incorreto , Quinolonas/farmacologia , Domínio Catalítico , Análise Mutacional de DNA , Integrase de HIV/química , Humanos , Modelos Moleculares , Estrutura Molecular
4.
J Biol Chem ; 283(44): 29904-11, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18728003

RESUMO

Indolopyridones are potent inhibitors of reverse transcriptase (RT) of the human immunodeficiency virus type 1 (HIV-1). Although the structure of these compounds differs from established nucleoside analogue RT inhibitors (NRTIs), previous studies suggest that the prototype compound INDOPY-1 may bind in close proximity to the polymerase active site. NRTI-associated mutations that are clustered around the active site confer decreased, e.g. M184V and Y115F, or increased, e.g. K65R, susceptibility to INDOPY-1. Here we have studied the underlying biochemical mechanism. RT enzymes containing the isolated mutations M184V and Y115F cause 2-3-fold increases in IC(50) values, while the combination of the two mutations causes a >15-fold increase. K65R can partially counteract these effects. Binding studies revealed that the M184V change reduces the affinity to INDOPY-1, while Y115F facilitates binding of the natural nucleotide substrate and the combined effects enhance the ability of the enzyme to discriminate against the inhibitor. Studies with other strategic mutations at residues Phe-61 and Ala-62, as well as the use of chemically modified templates shed further light on the putative binding site of the inhibitor and ternary complex formation. An abasic site residue at position n, i.e. opposite the 3'-end of the primer, prevents binding of INDOPY-1, while an abasic site at the adjacent position n+1 has no effect. Collectively, our findings provide strong evidence to suggest that INDOPY-1 can compete with natural deoxynucleoside triphosphates (dNTPs). We therefore propose to refer to members of this class of compounds as "nucleotide-competing RT inhibitors" (NcRTIs).


Assuntos
Transcriptase Reversa do HIV/genética , Mutação , Inibidores da Transcriptase Reversa/química , Sítios de Ligação , Domínio Catalítico , Primers do DNA/química , Dimerização , Relação Dose-Resposta a Droga , Escherichia coli/metabolismo , Concentração Inibidora 50 , Cinética , Modelos Químicos , Modelos Moleculares , Conformação Molecular , Ligação Proteica
5.
J Virol Methods ; 153(1): 7-15, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18640157

RESUMO

The high mutation rate of Human Immunodeficiency Virus (HIV) leads to the rapid derivation of compound-resistant virus strains and thus necessitates the identification and development of compounds with alternative mode of actions. MAPPIT (MAmmalian Protein-Protein Interaction Trap) is a highly efficient tool to study protein-protein interactions in intact human cells and is applied to study the dimerization process of the HIV reverse transcriptase complex. Highly specific signals for the p66/p51 and p66/p66 interactions could readily be detected. Specificity was established further by introducing mutations in either subunit. Treatment with efavirenz resulted in an increased MAPPIT signal, with an EC50 value of 64nM for the p66/p51 interaction, and allowed detection of the p51/p51 homodimerization, confirming the context-dependent asymmetric contribution of both subunits. These results show that MAPPIT can be used as a novel screening tool for anti-HIV compounds in intact human cells.


Assuntos
Transcriptase Reversa do HIV/metabolismo , HIV/fisiologia , Mapeamento de Interação de Proteínas/métodos , Alcinos , Benzoxazinas/farmacologia , Linhagem Celular , Ciclopropanos , Dimerização , Transcriptase Reversa do HIV/genética , Humanos , Concentração Inibidora 50 , Ligação Proteica , Subunidades Proteicas/metabolismo , Inibidores da Transcriptase Reversa/farmacologia
6.
J Virol Methods ; 151(1): 121-5, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18433887

RESUMO

The development of antiviral assays using an ATP/luminescence-based readout to profile antiviral compounds against the positive-strand RNA viruses: yellow fever virus (YFV), West Nile virus (WNV), Sindbis virus, and Coxsackie B virus, representing three virus families, is described. This assay readout is based upon the bioluminescent measurement of ATP in metabolically active cells. Antiviral efficacy was determined by measuring the ATP level in cells that were protected from the viral cytopathic effect (CPE) by the presence of antiviral compounds. The antiviral assay parameters were optimized and the assays were validated using a panel of different reference compounds to determine the intra- and inter-assay reproducibility. The signal-to-noise ratios for the yellow fever virus and West Nile virus assays were 7.5 and 36, respectively, comparing favorably with a signal-to-noise ratio of only 1.5 in the yellow fever virus neutral red dye uptake assay, an alternative readout for CPE inhibition. For Coxsackie B and Sindbis viruses, the signal-to-noise ratios were 40 and 50, respectively. These assays are robust, high-throughput, reproducible, and give much improved signal-to-noise ratios than those of dye uptake assays.


Assuntos
Trifosfato de Adenosina/metabolismo , Antivirais/farmacologia , Efeito Citopatogênico Viral/efeitos dos fármacos , Luminescência , Vírus de RNA/efeitos dos fármacos , Sindbis virus/efeitos dos fármacos , Animais , Chlorocebus aethiops , Enterovirus Humano B/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana/métodos , Vírus de RNA/classificação , Reprodutibilidade dos Testes , Células Vero , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus da Febre Amarela/efeitos dos fármacos
7.
J Virol ; 81(24): 13845-51, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17928344

RESUMO

The high incidence of cross-resistance between human immunodeficiency virus type 1 (HIV-1) protease inhibitors (PIs) limits their sequential use. This necessitates the development of PIs with a high genetic barrier and a broad spectrum of activity against PI-resistant HIV, such as tipranavir and darunavir (TMC114). We performed a surface plasmon resonance-based kinetic study to investigate the impact of PI resistance-associated mutations on the protease binding of five PIs used clinically: amprenavir, atazanavir, darunavir, lopinavir, and tipranavir. With wild-type protease, the binding affinity of darunavir was more than 100-fold higher than with the other PIs, due to a very slow dissociation rate. Consequently, the dissociative half-life of darunavir was much higher (>240 h) than that of the other PIs, including darunavir's structural analogue amprenavir. The influence of protease mutations on the binding kinetics was tested with five multidrug-resistant (MDR) proteases derived from clinical isolates harboring 10 to 14 PI resistance-associated mutations with a decreased susceptibility to various PIs. In general, all PIs bound to the MDR proteases with lower binding affinities, caused mainly by a faster dissociation rate. For amprenavir, atazanavir, lopinavir, and tipranavir, the decrease in affinity with MDR proteases resulted in reduced antiviral activity. For darunavir, however, a nearly 1,000-fold decrease in binding affinity did not translate into a weaker antiviral activity; a further decrease in affinity was required for the reduced antiviral effect. These observations provide a mechanistic explanation for darunavir's potent antiviral activity and high genetic barrier to the development of resistance.


Assuntos
Inibidores da Protease de HIV/metabolismo , Protease de HIV/genética , Protease de HIV/metabolismo , HIV-1/efeitos dos fármacos , Sulfonamidas/metabolismo , Sequência de Aminoácidos , Darunavir , Farmacorresistência Viral Múltipla , Farmacorresistência Viral , Inibidores da Protease de HIV/farmacologia , HIV-1/enzimologia , HIV-1/genética , Humanos , Cinética , Testes de Sensibilidade Microbiana/métodos , Dados de Sequência Molecular , Mutação , Sulfonamidas/farmacologia
8.
J Virol Methods ; 139(1): 17-23, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17034868

RESUMO

The use of targeting moieties is a new and exciting field of scientific research for facilitating the specific delivery of therapeutic agents in HIV-infected patients. The interaction of a potential targeting moiety with its ligand is a crucial factor in the evaluation of a targeted approach for chemotherapeutic intervention. Therefore, we have further characterized the interaction between a potential targeting agent, the monoclonal human antibody F105, and its ligand gp120, a glycoprotein expressed on the surface of HIV-1 infected cells. We demonstrate the specificity of binding and entry of F105 to infected cells. F105 was rapidly taken up into the cell and accumulated in the Golgi apparatus. Kinetic analysis of the F105-gp120 interaction revealed an equilibrium dissociation constant (K(D)) of 0.62 nM, compared with the gp120-CD4 interaction where the K(D) was determined at 35 nM. Consequently, F105 displayed a higher gp120 affinity. This was due to a slower dissociation as compared with the natural ligand. These data further underline the potential of monoclonal antibodies as targeting agents, and offer new insights into the possibility of F105 as a targeting moiety for the delivery of antiretroviral drugs to HIV-1 infected cells.


Assuntos
Síndrome da Imunodeficiência Adquirida/terapia , Anticorpos Monoclonais/metabolismo , Proteína gp120 do Envelope de HIV/imunologia , HIV-1 , Imunoglobulina G/metabolismo , Cadeias kappa de Imunoglobulina/metabolismo , Anticorpos Monoclonais/uso terapêutico , Humanos
9.
J Immunol ; 177(9): 6227-37, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17056552

RESUMO

To gain insights into the molecular mechanisms underlying early host responses to HIV in the CD4(+) T cell target population, we examined gene expression in CD4(+) T cells isolated 24 h after ex vivo HIV infection of lymphocyte aggregate cultures derived from human tonsils. Gene profiling showed a distinct up-regulation of genes related to immune response and response to virus, notably of IFN-stimulated genes (ISGs), irrespective of the coreceptor tropism of the virus. This mostly IFN-alpha-dependent gene signature suggested the involvement of plasmacytoid dendritic cells, a principal component of the antiviral immune response. Indeed, depletion of plasmacytoid dendritic cells before HIV inoculation abrogated transcriptional up-regulation of several ISGs and resulted in increased levels of HIV replication. Treatment with a blocking anti-IFN-alphaR Ab yielded increased HIV replication; conversely, HIV replication was decreased in pDC-depleted cultures treated with IFN-alpha. Among up-regulated ISGs was also TRAIL, indicating a potential role of the IFN signature in apoptosis. However, a blocking anti-TRAIL Ab did not abrogate apoptosis of CD4(+) T cells in CXCR4-tropic HIV-infected cultures, suggesting the involvement of pathways other than TRAIL mediated. We conclude that acute HIV infection of lymphoid tissue results in up-regulation of ISGs in CD4(+) T cells, which induces an anti-HIV state but not apoptosis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , HIV/fisiologia , Interferon-alfa/fisiologia , Anticorpos Bloqueadores/farmacologia , Apoptose/efeitos dos fármacos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Células Dendríticas/imunologia , Perfilação da Expressão Gênica , HIV/efeitos dos fármacos , Humanos , Imunidade/genética , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/farmacologia , Interferons/antagonistas & inibidores , Interferons/farmacologia , Tonsila Palatina/virologia , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Regulação para Cima , Replicação Viral/efeitos dos fármacos
10.
Virology ; 356(1-2): 79-94, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16926043

RESUMO

In order to identify cellular genes which interfere with HIV-1 replication in monocyte-derived macrophages (MAC), cells were stimulated with interferon (IFN) or lipopolysaccharide (LPS) leading to a pronounced inhibition of HIV-1 infection in these cells, and the resulting gene expression was analyzed. Using the microarray technology we identified a gene named Stimulated Trans-Acting Factor of 50 kDa (Staf50), which is known to repress the activity of the HIV-1 LTR. Analysis of the Staf50 expression by real-time PCR showed an overexpression in IFNalpha (up to 20-fold) and LPS (up to 10-fold)-stimulated MAC as well as in infected cells (up to 3-fold). For stable overexpression, 293 T cells and primary macrophages were transduced with Staf50-IRES-GFP bicistronic pseudotype viruses. After transduction, 293 T CD4/CCR5 and MAC were infected with HIV-1, and virus replication was monitored by p24 ELISA. Overexpression of Staf50 inhibited the HIV-1 infection between 50% and 90% in 293 T CD4/CCR5 as well as in MAC. Our findings suggest that host genetic effects in combination with viral properties determine the susceptibility of an appropriate target cell for HIV-1 infection as well as the replication potential of the virus in the cell resulting in an overall productive infection.


Assuntos
HIV-1/fisiologia , Macrófagos/virologia , Proteínas Repressoras/metabolismo , Proteínas Repressoras/farmacologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/farmacologia , Regulação para Cima , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Perfilação da Expressão Gênica , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Humanos , Interferon-alfa/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Macrófagos/metabolismo , Antígenos de Histocompatibilidade Menor , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Proteínas com Motivo Tripartido
11.
Antiviral Res ; 65(1): 35-43, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15652969

RESUMO

New antiviral drugs are needed for the treatment of cytomegalovirus (CMV) infections, particularly in immunocompromised patients. These studies evaluated the in vitro and in vivo activity of the non-nucleosidic CMV inhibitor, BAY 38-4766, against guinea pig cytomegalovirus (GPCMV). Plaque reduction assays indicated that BAY 38-4766 was active against GPCMV, with an IC(50) of 0.5muM. Yield reduction assays demonstrated an ED(90) and ED(99) of 0.4 and 0.6muM, respectively, of BAY 38-4766 against GPCMV. Guinea pigs tolerated oral administration of 50mg/kg/day of BAY 38-4766 without evidence of biochemical or hematologic toxicity. Plasma concentrations of BAY 38-4766 were high following oral dosing, with a mean peak level at 1-h post-dose of 26.7mg/ml (n=6; range, 17.8-35.4). Treatment with BAY 38-4766 reduced both viremia and DNAemia, as determined by a real-time PCR assay, following GPCMV infection of cyclophosphamide-immunosuppressed strain 2 guinea pigs (p<0.05, Mann-Whitney test). BAY 38-4766 also reduced mortality following lethal GPCMV challenge in immunosuppressed Hartley guinea pigs, from 83% (20/24) in placebo-treated guinea pigs, to 17% (4/24) in BAY 38-4766-treated animals (p<0.0001, Fisher's exact test). Mortality differences were accompanied by reduction in DNAemia in Hartley guinea pigs. Based upon its favorable safety, pharmacokinetic, and therapeutic profiles, BAY 38-4766 warrants further investigation in the GPCMV model.


Assuntos
Antivirais/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/mortalidade , Citomegalovirus/efeitos dos fármacos , Hospedeiro Imunocomprometido , Naftalenossulfonatos/uso terapêutico , Animais , Antivirais/efeitos adversos , Antivirais/farmacocinética , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Modelos Animais de Doenças , Farmacorresistência Viral , Cobaias , Naftalenossulfonatos/efeitos adversos , Naftalenossulfonatos/farmacocinética , Resultado do Tratamento
12.
Curr Opin Pharmacol ; 4(5): 453-64, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15351349

RESUMO

Antiviral drugs against DNA viruses are widely used for the management of diseases caused by infections with the Herpes viruses and have recently been introduced for Hepatitis B. There are also several emerging treatments (i.e. those that are in clinical development) and novel treatments that are still in the preclinical phase. Although the majority of emerging drugs are nucleoside analogues, there is a trend towards the development of non-nucleosidic drugs with unique mechanisms of action, in the hope that efficacy will be maximised and drug resistance and viral rebound minimised.


Assuntos
Antivirais/uso terapêutico , Hepatite B/tratamento farmacológico , Infecções por Herpesviridae/tratamento farmacológico , Antivirais/química , Ensaios Clínicos como Assunto , Infecções por Citomegalovirus/tratamento farmacológico , Herpes Simples/tratamento farmacológico , Herpes Zoster/tratamento farmacológico , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA