Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Chem Rev ; 121(6): 3238-3270, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33410674

RESUMO

Drug resistance is prevalent across many diseases, rendering therapies ineffective with severe financial and health consequences. Rather than accepting resistance after the fact, proactive strategies need to be incorporated into the drug design and development process to minimize the impact of drug resistance. These strategies can be derived from our experience with viral disease targets where multiple generations of drugs had to be developed to combat resistance and avoid antiviral failure. Significant efforts including experimental and computational structural biology, medicinal chemistry, and machine learning have focused on understanding the mechanisms and structural basis of resistance against direct-acting antiviral (DAA) drugs. Integrated methods show promise for being predictive of resistance and potency. In this review, we give an overview of this research for human immunodeficiency virus type 1, hepatitis C virus, and influenza virus and the lessons learned from resistance mechanisms of DAAs. These lessons translate into rational strategies to avoid resistance in drug design, which can be generalized and applied beyond viral targets. While resistance may not be completely avoidable, rational drug design can and should incorporate strategies at the outset of drug development to decrease the prevalence of drug resistance.


Assuntos
Antivirais/química , Inibidores Enzimáticos/química , Preparações Farmacêuticas/química , Proteínas Virais/química , Viroses/tratamento farmacológico , Antivirais/metabolismo , Antivirais/farmacologia , Biologia Computacional , Desenho de Fármacos , Farmacorresistência Viral , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , HIV-1/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Humanos , Aprendizado de Máquina , Mutação , Orthomyxoviridae/efeitos dos fármacos , Preparações Farmacêuticas/metabolismo , Ligação Proteica , Transdução de Sinais , Relação Estrutura-Atividade
2.
Bioorg Chem ; 131: 106269, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36446201

RESUMO

Viruses from the Flavivirus genus infect millions of people worldwide and cause severe diseases, including recent epidemics of dengue virus (DENV), and Zika virus (ZIKV). There is currently no antiviral treatment against flavivirus infections, despite considerable efforts to develop inhibitors against essential viral enzymes including NS2B/NS3 protease. Targeting the flavivirus NS2B/NS3 protease proved to be challenging because of the conformational dynamics, topology, and electrostatic properties of the active site. Here, we report the identification of quinoxaline-based allosteric inhibitors by fragment-based drug discovery approach as a promising new drug-like scaffold to target the NS2B/NS3 protease. Enzymatic assays and mutational analysis of the allosteric site in ZIKV NS2B/NS3 protease support noncompetitive inhibition mechanism as well as engineered DENV protease construct indicating the compounds likely compete with the NS2B cofactor for binding to the protease domain. Furthermore, antiviral activity confirmed the therapeutic potential of this new inhibitor scaffold.


Assuntos
Flavivirus , Infecção por Zika virus , Zika virus , Humanos , Flavivirus/química , Flavivirus/metabolismo , Zika virus/metabolismo , Peptídeo Hidrolases , Quinoxalinas/farmacologia , Proteínas não Estruturais Virais , Serina Endopeptidases/metabolismo , Inibidores de Proteases/farmacologia , Antivirais/química
3.
Biochemistry ; 60(39): 2925-2931, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34506130

RESUMO

Rupintrivir targets the 3C cysteine proteases of the picornaviridae family, which includes rhinoviruses and enteroviruses that cause a range of human diseases. Despite being a pan-3C protease inhibitor, rupintrivir activity is extremely weak against the homologous 3C-like protease of SARS-CoV-2. In this study, the crystal structures of rupintrivir were determined bound to enterovirus 68 (EV68) 3C protease and the 3C-like main protease (Mpro) from SARS-CoV-2. While the EV68 3C protease-rupintrivir structure was similar to previously determined complexes with other picornavirus 3C proteases, rupintrivir bound in a unique conformation to the active site of SARS-CoV-2 Mpro splitting the catalytic cysteine and histidine residues. This bifurcation of the catalytic dyad may provide a novel approach for inhibiting cysteine proteases.


Assuntos
Antivirais/metabolismo , Proteases 3C de Coronavírus/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Isoxazóis/metabolismo , Fenilalanina/análogos & derivados , Pirrolidinonas/metabolismo , SARS-CoV-2/enzimologia , Valina/análogos & derivados , Antivirais/química , Domínio Catalítico , Proteases 3C de Coronavírus/antagonistas & inibidores , Proteases 3C de Coronavírus/química , Cristalografia por Raios X , Inibidores de Cisteína Proteinase/química , Enterovirus Humano D/enzimologia , Ligação de Hidrogênio , Isoxazóis/química , Fenilalanina/química , Fenilalanina/metabolismo , Ligação Proteica , Pirrolidinonas/química , Eletricidade Estática , Valina/química , Valina/metabolismo
4.
Biochemistry ; 58(35): 3711-3726, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31386353

RESUMO

Protease inhibitors have the highest potency among antiviral therapies against HIV-1 infections, yet the virus can evolve resistance. Darunavir (DRV), currently the most potent Food and Drug Administration-approved protease inhibitor, retains potency against single-site mutations. However, complex combinations of mutations can confer resistance to DRV. While the interdependence between mutations within HIV-1 protease is key for inhibitor potency, the molecular mechanisms that underlie this control remain largely unknown. In this study, we investigated the interdependence between the L89V and L90M mutations and their effects on DRV binding. These two mutations have been reported to be positively correlated with one another in HIV-1 patient-derived protease isolates, with the presence of one mutation making the probability of the occurrence of the second mutation more likely. The focus of our investigation is a patient-derived isolate, with 24 mutations that we call "KY"; this variant includes the L89V and L90M mutations. Three additional KY variants with back-mutations, KY(V89L), KY(M90L), and the KY(V89L/M90L) double mutation, were used to experimentally assess the individual and combined effects of these mutations on DRV inhibition and substrate processing. The enzymatic assays revealed that the KY(V89L) variant, with methionine at residue 90, is highly resistant, but its catalytic function is compromised. When a leucine to valine mutation at residue 89 is present simultaneously with the L90M mutation, a rescue of catalytic efficiency is observed. Molecular dynamics simulations of these DRV-bound protease variants reveal how the L90M mutation induces structural changes throughout the enzyme that undermine the binding interactions.


Assuntos
Substituição de Aminoácidos/fisiologia , Farmacorresistência Viral/genética , Epistasia Genética/genética , Protease de HIV/genética , Substituição de Aminoácidos/genética , Domínio Catalítico , Cristalografia por Raios X , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Protease de HIV/química , Protease de HIV/metabolismo , Inibidores da Protease de HIV/farmacologia , Inibidores da Protease de HIV/uso terapêutico , HIV-1/enzimologia , HIV-1/genética , Humanos , Leucina/genética , Metionina/genética , Modelos Moleculares , Simulação de Dinâmica Molecular , Mutação de Sentido Incorreto/fisiologia , Ligação Proteica , Desnaturação Proteica , Valina/genética
5.
J Am Chem Soc ; 136(34): 11956-63, 2014 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25091085

RESUMO

HIV-1 protease inhibitors are part of the highly active antiretroviral therapy effectively used in the treatment of HIV infection and AIDS. Darunavir (DRV) is the most potent of these inhibitors, soliciting drug resistance only when a complex combination of mutations occur both inside and outside the protease active site. With few exceptions, the role of mutations outside the active site in conferring resistance remains largely elusive. Through a series of DRV-protease complex crystal structures, inhibition assays, and molecular dynamics simulations, we find that single and double site mutations outside the active site often associated with DRV resistance alter the structure and dynamic ensemble of HIV-1 protease active site. These alterations correlate with the observed inhibitor binding affinities for the mutants, and suggest a network hypothesis on how the effect of distal mutations are propagated to pivotal residues at the active site and may contribute to conferring drug resistance.


Assuntos
Farmacorresistência Viral/genética , Inibidores da Protease de HIV/farmacologia , Protease de HIV/química , HIV-1/enzimologia , Mutação , Sulfonamidas/farmacologia , Sítios de Ligação , Darunavir , Protease de HIV/genética , Protease de HIV/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica
6.
J Virol ; 87(8): 4176-84, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23365446

RESUMO

Drug resistance occurs through a series of subtle changes that maintain substrate recognition but no longer permit inhibitor binding. In HIV-1 protease, mutations at I50 are associated with such subtle changes that confer differential resistance to specific inhibitors. Residue I50 is located at the protease flap tips, closing the active site upon ligand binding. Under selective drug pressure, I50V/L substitutions emerge in patients, compromising drug susceptibility and leading to treatment failure. The I50V substitution is often associated with amprenavir (APV) and darunavir (DRV) resistance, while the I50L substitution is observed in patients failing atazanavir (ATV) therapy. To explain how APV, DRV, and ATV susceptibility are influenced by mutations at residue 50 in HIV-1 protease, structural and binding thermodynamics studies were carried out on I50V/L-substituted protease variants in the compensatory mutation A71V background. Reduced affinity to both I50V/A71V and I50L/A71V double mutants is largely due to decreased binding entropy, which is compensated for by enhanced enthalpy for ATV binding to I50V variants and APV binding to I50L variants, leading to hypersusceptibility in these two cases. Analysis of the crystal structures showed that the substitutions at residue 50 affect how APV, DRV, and ATV bind the protease with altered van der Waals interactions and that the selection of I50V versus I50L is greatly influenced by the chemical moieties at the P1 position for APV/DRV and the P2 position for ATV. Thus, the varied inhibitor susceptibilities of I50V/L protease variants are largely a direct consequence of the interdependent changes in protease inhibitor interactions.


Assuntos
Fármacos Anti-HIV/farmacologia , Farmacorresistência Viral , Protease de HIV/química , HIV-1/efeitos dos fármacos , Mutação de Sentido Incorreto , Sulfato de Atazanavir , Carbamatos/farmacologia , Cristalografia por Raios X , Darunavir , Furanos , Protease de HIV/genética , HIV-1/genética , Humanos , Cinética , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Oligopeptídeos/farmacologia , Mutação Puntual , Ligação Proteica , Conformação Proteica , Piridinas/farmacologia , Sulfonamidas/farmacologia , Termodinâmica
7.
Eur J Med Chem ; 257: 115501, 2023 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-37244161

RESUMO

Protease inhibitors are the most potent antivirals against HIV-1, but they still lose efficacy against resistant variants. Improving the resistance profile is key to developing more robust inhibitors, which may be promising candidates for simplified next-generation antiretroviral therapies. In this study, we explored analogs of darunavir with a P1 phosphonate modification in combination with increasing size of the P1' hydrophobic group and various P2' moieties to improve potency against resistant variants. The phosphonate moiety substantially improved potency against highly mutated and resistant HIV-1 protease variants, but only when combined with more hydrophobic moieties at the P1' and P2' positions. Phosphonate analogs with a larger hydrophobic P1' moiety maintained excellent antiviral potency against a panel of highly resistant HIV-1 variants, with significantly improved resistance profiles. The cocrystal structures indicate that the phosphonate moiety makes extensive hydrophobic interactions with the protease, especially with the flap residues. Many residues involved in these protease-inhibitor interactions are conserved, enabling the inhibitors to maintain potency against highly resistant variants. These results highlight the need to balance inhibitor physicochemical properties by simultaneous modification of chemical groups to further improve resistance profiles.


Assuntos
Inibidores da Protease de HIV , HIV-1 , Inibidores da Protease de HIV/farmacologia , Inibidores da Protease de HIV/química , Darunavir/farmacologia , Peptídeo Hidrolases , Protease de HIV/genética , Cristalografia por Raios X
8.
Elife ; 122023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36920025

RESUMO

Darunavir (DRV) is exceptional among potent HIV-1 protease inhibitors (PIs) in high drug concentrations that are achieved in vivo. Little is known about the de novo resistance pathway for DRV. We selected for resistance to high drug concentrations against 10 PIs and their structural precursor DRV. Mutations accumulated through two pathways (anchored by protease mutations I50V or I84V). Small changes in the inhibitor P1'-equivalent position led to preferential use of one pathway over the other. Changes in the inhibitor P2'-equivalent position determined differences in potency that were retained in the resistant viruses and that impacted the selected mutations. Viral variants from the two pathways showed differential selection of compensatory mutations in Gag cleavage sites. These results reveal the high level of selective pressure that is attainable with fifth-generation PIs and how features of the inhibitor affect both the resistance pathway and the residual potency in the face of resistance.


Assuntos
Infecções por HIV , Inibidores da Protease de HIV , HIV-1 , Humanos , Inibidores da Protease de HIV/química , HIV-1/genética , Darunavir/farmacologia , Darunavir/uso terapêutico , Mutação , Farmacorresistência Viral/genética , Infecções por HIV/tratamento farmacológico
9.
J Virol ; 84(19): 9995-10003, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660190

RESUMO

The majority of HIV-1 infections around the world result from non-B clade HIV-1 strains. The CRF01_AE (AE) strain is seen principally in Southeast Asia. AE protease differs by approximately 10% in amino acid sequence from clade B protease and carries several naturally occurring polymorphisms that are associated with drug resistance in clade B. AE protease has been observed to develop resistance through a nonactive-site N88S mutation in response to nelfinavir (NFV) therapy, whereas clade B protease develops both the active-site mutation D30N and the nonactive-site mutation N88D. Structural and biochemical studies were carried out with wild-type and NFV-resistant clade B and AE protease variants. The relationship between clade-specific sequence variations and pathways to inhibitor resistance was also assessed. AE protease has a lower catalytic turnover rate than clade B protease, and it also has weaker affinity for both NFV and darunavir (DRV). This weaker affinity may lead to the nonactive-site N88S variant in AE, which exhibits significantly decreased affinity for both NFV and DRV. The D30N/N88D mutations in clade B resulted in a significant loss of affinity for NFV and, to a lesser extent, for DRV. A comparison of crystal structures of AE protease shows significant structural rearrangement in the flap hinge region compared with those of clade B protease and suggests insights into the alternative pathways to NFV resistance. In combination, our studies show that sequence polymorphisms within clades can alter protease activity and inhibitor binding and are capable of altering the pathway to inhibitor resistance.


Assuntos
Inibidores da Protease de HIV/farmacologia , Protease de HIV/genética , HIV-1/enzimologia , HIV-1/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Domínio Catalítico/genética , Cristalografia por Raios X , Darunavir , Farmacorresistência Viral/genética , Genes Virais , Protease de HIV/química , Protease de HIV/metabolismo , HIV-1/classificação , HIV-1/efeitos dos fármacos , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Nelfinavir/farmacologia , Polimorfismo Genético , Conformação Proteica , Homologia de Sequência de Aminoácidos , Sulfonamidas/farmacologia , Termodinâmica
10.
ACS Chem Biol ; 16(3): 529-538, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33619959

RESUMO

Human T-cell lymphotropic virus type 1 (HTLV-1) is a retrovirus that can cause severe paralytic neurologic disease and immune disorders as well as cancer. An estimated 20 million people worldwide are infected with HTLV-1, with prevalence reaching 30% in some parts of the world. In stark contrast to HIV-1, no direct acting antivirals (DAAs) exist against HTLV-1. The aspartyl protease of HTLV-1 is a dimer similar to that of HIV-1 and processes the viral polyprotein to permit viral maturation. We report that the FDA-approved HIV-1 protease inhibitor darunavir (DRV) inhibits the enzyme with 0.8 µM potency and provides a scaffold for drug design against HTLV-1. Analogs of DRV that we designed and synthesized achieved submicromolar inhibition against HTLV-1 protease and inhibited Gag processing in viral maturation assays and in a chronically HTLV-1 infected cell line. Cocrystal structures of these inhibitors with HTLV-1 protease highlight opportunities for future inhibitor design. Our results show promise toward developing highly potent HTLV-1 protease inhibitors as therapeutic agents against HTLV-1 infections.


Assuntos
Antivirais/química , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Darunavir/análogos & derivados , Vírus Linfotrópico T Tipo 1 Humano/efeitos dos fármacos , Inibidores de Proteases/química , Sequência de Aminoácidos , Antivirais/farmacologia , Ácido Aspártico Endopeptidases/química , Ácido Aspártico Endopeptidases/genética , Darunavir/farmacologia , Descoberta de Drogas , Escherichia coli/genética , Humanos , Simulação de Dinâmica Molecular , Estrutura Molecular , Terapia de Alvo Molecular , Inibidores de Proteases/farmacologia , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA