Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antimicrob Agents Chemother ; 68(4): e0137323, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38380945

RESUMEN

Protease inhibitors (PIs) remain an important component of antiretroviral therapy for the treatment of HIV-1 infection due to their high genetic barrier to resistance development. Nevertheless, the two most commonly prescribed HIV PIs, atazanavir and darunavir, still require co-administration with a pharmacokinetic boosting agent to maintain sufficient drug plasma levels which can lead to undesirable drug-drug interactions. Herein, we describe GS-9770, a novel investigational non-peptidomimetic HIV PI with unboosted once-daily oral dosing potential due to improvements in its metabolic stability and its pharmacokinetic properties in preclinical animal species. This compound demonstrates potent inhibitory activity and high on-target selectivity for recombinant HIV-1 protease versus other aspartic proteases tested. In cell culture, GS-9770 inhibits Gag polyprotein cleavage and shows nanomolar anti-HIV-1 potency in primary human cells permissive to HIV-1 infection and against a broad range of HIV subtypes. GS-9770 demonstrates an improved resistance profile against a panel of patient-derived HIV-1 isolates with resistance to atazanavir and darunavir. In resistance selection experiments, GS-9770 prevented the emergence of breakthrough HIV-1 variants at all fixed drug concentrations tested and required multiple protease substitutions to enable outgrowth of virus exposed to escalating concentrations of GS-9770. This compound also remained fully active against viruses resistant to drugs from other antiviral classes and showed no in vitro antagonism when combined pairwise with drugs from other antiretroviral classes. Collectively, these preclinical data identify GS-9770 as a potent, non-peptidomimetic once-daily oral HIV PI with potential to overcome the persistent requirement for pharmacological boosting with this class of antiretroviral agents.


Asunto(s)
Infecciones por VIH , Inhibidores de la Proteasa del VIH , VIH-1 , Humanos , Inhibidores de la Proteasa del VIH/farmacología , Inhibidores de la Proteasa del VIH/uso terapéutico , Darunavir/farmacología , Darunavir/uso terapéutico , Sulfato de Atazanavir/farmacología , Sulfato de Atazanavir/uso terapéutico , Farmacorresistencia Viral , VIH-1/genética , Antirretrovirales/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Proteasa del VIH/genética , Proteasa del VIH/metabolismo
2.
ACS Infect Dis ; 9(10): 1918-1931, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37728236

RESUMEN

A collaborative, open-science team undertook discovery of novel small molecule inhibitors of the SARS-CoV-2 nsp16-nsp10 2'-O-methyltransferase using a high throughput screening approach with the potential to reveal new inhibition strategies. This screen yielded compound 5a, a ligand possessing an electron-deficient double bond, as an inhibitor of SARS-CoV-2 nsp16 activity. Surprisingly, X-ray crystal structures revealed that 5a covalently binds within a previously unrecognized cryptic pocket near the S-adenosylmethionine binding cleft in a manner that prevents occupation by S-adenosylmethionine. Using a multidisciplinary approach, we examined the mechanism of binding of compound 5a to the nsp16 cryptic pocket and developed 5a derivatives that inhibited nsp16 activity and murine hepatitis virus replication in rat lung epithelial cells but proved cytotoxic to cell lines canonically used to examine SARS-CoV-2 infection. Our study reveals the druggability of this newly discovered SARS-CoV-2 nsp16 cryptic pocket, provides novel tool compounds to explore the site, and suggests a new approach for discovery of nsp16 inhibition-based pan-coronavirus therapeutics through structure-guided drug design.


Asunto(s)
COVID-19 , SARS-CoV-2 , Ratones , Ratas , Animales , SARS-CoV-2/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Metiltransferasas
3.
Nat Rev Drug Discov ; 22(7): 585-603, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37173515

RESUMEN

During the coronavirus disease 2019 (COVID-19) pandemic, a wave of rapid and collaborative drug discovery efforts took place in academia and industry, culminating in several therapeutics being discovered, approved and deployed in a 2-year time frame. This article summarizes the collective experience of several pharmaceutical companies and academic collaborations that were active in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antiviral discovery. We outline our opinions and experiences on key stages in the small-molecule drug discovery process: target selection, medicinal chemistry, antiviral assays, animal efficacy and attempts to pre-empt resistance. We propose strategies that could accelerate future efforts and argue that a key bottleneck is the lack of quality chemical probes around understudied viral targets, which would serve as a starting point for drug discovery. Considering the small size of the viral proteome, comprehensively building an arsenal of probes for proteins in viruses of pandemic concern is a worthwhile and tractable challenge for the community.


Asunto(s)
COVID-19 , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , SARS-CoV-2 , Descubrimiento de Drogas , Pandemias
4.
PLoS One ; 17(12): e0271145, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36477212

RESUMEN

Chronic hepatitis B (CHB) is a global health care challenge and a major cause of liver disease. To find new therapeutic avenues with a potential to functionally cure chronic Hepatitis B virus (HBV) infection, we performed a focused screen of epigenetic modifiers to identify potential inhibitors of replication or gene expression. From this work we identified isonicotinic acid inhibitors of the histone lysine demethylase 5 (KDM5) with potent anti-HBV activity. To enhance the cellular permeability and liver accumulation of the most potent KDM5 inhibitor identified (GS-080) an ester prodrug was developed (GS-5801) that resulted in improved bioavailability and liver exposure as well as an increased H3K4me3:H3 ratio on chromatin. GS-5801 treatment of HBV-infected primary human hepatocytes reduced the levels of HBV RNA, DNA and antigen. Evaluation of GS-5801 antiviral activity in a humanized mouse model of HBV infection, however, did not result in antiviral efficacy, despite achieving pharmacodynamic levels of H3K4me3:H3 predicted to be efficacious from the in vitro model. Here we discuss potential reasons for the disconnect between in vitro and in vivo efficacy, which highlight the translational difficulties of epigenetic targets for viral diseases.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B Crónica , Humanos , Animales , Ratones , Antivirales/farmacología , Hepatitis B Crónica/tratamiento farmacológico , Epigenómica
5.
Antivir Ther ; 27(2): 13596535211067598, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35499178

RESUMEN

John Martin's untimely death in March 2021 was a huge loss for us personally, Gilead Sciences, the company he built over 30 years and the scientific community concerned with antiviral therapies. We wish to honor John's legacy by retelling the discovery and history of Tamiflu and his contributions to it. Without his vision, persistence, and keen eye for opportunities, Tamiflu would not exist and Gilead's path would not have been the same. His strategic thinking around the first oral flu drug is still quite relevant today, when we are still in the SARS-CoV-2 pandemic. John explained it simply in an interview with the Science History Institute in May 2020: "…most of my colleagues, we travel with Tamiflu when we go internationally, because it works for treatment and prevention, and hopefully, there will be a solution like that, eventually, for the Covid virus in addition to vaccines. Most people will get a flu vaccine every year, but there is still disease, we need a pill for treatment and prevention.".


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Gripe Humana , Humanos , Gripe Humana/tratamiento farmacológico , Gripe Humana/prevención & control , Oseltamivir/farmacología , Oseltamivir/uso terapéutico , SARS-CoV-2
6.
J Biol Chem ; 297(4): 101218, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34562452

RESUMEN

The SARS-CoV-2 replication-transcription complex is an assembly of nonstructural viral proteins that collectively act to reproduce the viral genome and generate mRNA transcripts. While the structures of the individual proteins involved are known, how they assemble into a functioning superstructure is not. Applying molecular modeling tools, including protein-protein docking, to the available structures of nsp7-nsp16 and the nucleocapsid, we have constructed an atomistic model of how these proteins associate. Our principal finding is that the complex is hexameric, centered on nsp15. The nsp15 hexamer is capped on two faces by trimers of nsp14/nsp16/(nsp10)2, which then recruit six nsp12/nsp7/(nsp8)2 polymerase subunits to the complex. To this, six subunits of nsp13 are arranged around the superstructure, but not evenly distributed. Polymerase subunits that coordinate dimers of nsp13 are capable of binding the nucleocapsid, which positions the 5'-UTR TRS-L RNA over the polymerase active site, a state distinguishing transcription from replication. Analysis of the viral RNA path through the complex indicates the dsRNA that exits the polymerase passes over the nsp14 exonuclease and nsp15 endonuclease sites before being unwound by a convergence of zinc fingers from nsp10 and nsp14. The template strand is then directed away from the complex, while the nascent strand is directed to the sites responsible for mRNA capping. The model presents a cohesive picture of the multiple functions of the coronavirus replication-transcription complex and addresses fundamental questions related to proofreading, template switching, mRNA capping, and the role of the endonuclease.


Asunto(s)
Endorribonucleasas/metabolismo , Modelos Moleculares , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/metabolismo , Sitios de Unión , COVID-19/patología , COVID-19/virología , Dimerización , Endorribonucleasas/química , Endorribonucleasas/genética , Humanos , Simulación del Acoplamiento Molecular , Estructura Cuaternaria de Proteína , ARN Bicatenario/química , ARN Bicatenario/metabolismo , SARS-CoV-2/aislamiento & purificación , Transcripción Genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Replicación Viral
7.
J Chem Inf Model ; 60(7): 3489-3498, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32539379

RESUMEN

A tremendous research and development effort was exerted toward combating chronic hepatitis C, ultimately leading to curative oral treatments, all of which are targeting viral proteins. Despite the advantage of numerous targets allowing for broad hepatitis C virus (HCV) genotype coverage, the only host target inhibitors that advanced into clinical development were Cyclosporin A based cyclophilin inhibitors. While cyclosporin-based molecules typically require a fermentation process, Gilead successfully pursued a fully synthetic, oral program based on Sanglifehrin A. The drug discovery process, though greatly helped by facile crystallography, was still hampered by the limitations in the accuracy of predictive computational methods for prioritizing compound ideas. Recent advances in accuracy and speed of free energy perturbation (FEP) methods, however, are attractive for prioritizing and derisking synthetically challenging molecules and potentially could have had a significant impact on the speed of the development of this program. Here in our simulated prospective study, the binding free energies of 26 macrocyclic cyclophilin inhibitors were blindly predicted using FEP+ to test this hypothesis. The predictions had a low mean unsigned error (MUE) (1.1 kcal/mol) and accurately reproduced many design decisions from the program, suggesting that FEP+ has the potential to drive synthetic chemistry efforts by more accurately ranking compounds with nonintuitive structure-activity relationships (SARs).


Asunto(s)
Descubrimiento de Drogas , Entropía , Estudios Prospectivos , Relación Estructura-Actividad , Termodinámica
8.
Antiviral Res ; 169: 104538, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31226346

RESUMEN

We describe here the anti-HBV activity of natural and synthetic retinoids in primary human hepatocytes (PHHs). The most potent compounds inhibited HBsAg, HBeAg, viral RNA and DNA production by HBV infected cells with EC50 values ranging from 0.4 to 2.6 µM. The activity was independent of PHH donor and HBV genotype used in testing. 13-cis retinoic acid (Accutane) was selected for further evaluation in the PXB chimeric mouse model of HBV infection at doses allowing to achieve Accutane peak serum concentrations near its antiviral EC90 and exposures ∼5-fold higher than a typical clinical dose. While these supraclinical exposures of 100 mg/kg/day were well-tolerated by regular Balb/c mice, PXB mice were more sensitive and even a lower those of 60 mg/kg/day led to significant weight loss. Despite dosing at this maximal tolerated dose for 28 days, Accutane failed to show any anti-HBV activity. RAR target engagement was verified using transcriptome analysis of liver samples from treated versus vehicle groups. However, gene expression changes in PXB liver samples were vastly muted when compared to the in vitro PHH system. When comparing transcriptional changes associated with the conditioning of fresh hepatocytes toward enabling HBV infection, we also observed a large number of changes. Noticeably, a significant number of genes that were up- or down-regulated by the conditioning process were down- or up-regulated by HBV infected PHH treatment with Accutane, respectively. While the lack of efficacy in the PXB model may have many explanations, the observed, opposing transcriptional changes upon conditioning PHH and treating these cultured, HBV-infected PHH with Accutane allow for the possibility that the PHH system may yield artificial anti-HBV hits.


Asunto(s)
Antivirales/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Hepatocitos/virología , Retinoides/farmacología , Animales , Antivirales/sangre , Supervivencia Celular/efectos de los fármacos , ADN Viral/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Expresión Génica/efectos de los fármacos , Hepatitis B/virología , Antígenos de Superficie de la Hepatitis B/efectos de los fármacos , Antígenos e de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Hepatocitos/metabolismo , Humanos , Isotretinoína/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , ARN Viral/metabolismo , Retinoides/sangre , Regulación hacia Arriba , Replicación Viral/efectos de los fármacos
9.
J Med Chem ; 61(21): 9473-9499, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30074795

RESUMEN

Cyclophilins are a family of peptidyl-prolyl isomerases that are implicated in a wide range of diseases including hepatitis C. Our aim was to discover through total synthesis an orally bioavailable, non-immunosuppressive cyclophilin (Cyp) inhibitor with potent anti-hepatitis C virus (HCV) activity that could serve as part of an all oral antiviral combination therapy. An initial lead 2 derived from the sanglifehrin A macrocycle was optimized using structure based design to produce a potent and orally bioavailable inhibitor 3. The macrocycle ring size was reduced by one atom, and an internal hydrogen bond drove improved permeability and drug-like properties. 3 demonstrates potent Cyp inhibition ( Kd = 5 nM), potent anti-HCV 2a activity (EC50 = 98 nM), and high oral bioavailability in rat (100%) and dog (55%). The synthetic accessibility and properties of 3 support its potential as an anti-HCV agent and for interrogating the role of Cyp inhibition in a variety of diseases.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/farmacocinética , Administración Oral , Antivirales/administración & dosificación , Antivirales/química , Antivirales/farmacocinética , Antivirales/farmacología , Disponibilidad Biológica , Línea Celular , Ciclofilinas/química , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Hepacivirus/efectos de los fármacos , Lactonas/administración & dosificación , Lactonas/química , Lactonas/farmacocinética , Lactonas/farmacología , Modelos Moleculares , Conformación Proteica , Compuestos de Espiro/administración & dosificación , Compuestos de Espiro/química , Compuestos de Espiro/farmacocinética , Compuestos de Espiro/farmacología
10.
J Med Chem ; 60(3): 1000-1017, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28075591

RESUMEN

Cyclophilin inhibition has been a target for the treatment of hepatitis C and other diseases, but the generation of potent, drug-like molecules through chemical synthesis has been challenging. In this study, a set of macrocyclic cyclophilin inhibitors was synthesized based on the core structure of the natural product sanglifehrin A. Initial compound optimization identified the valine-m-tyrosine-piperazic acid tripeptide (Val-m-Tyr-Pip) in the sanglifehrin core, stereocenters at C14 and C15, and the hydroxyl group of the m-tyrosine (m-Tyr) residue as key contributors to compound potency. Replacing the C18-C21 diene unit of sanglifehrin with a styryl group led to potent compounds that displayed a novel binding mode in which the styrene moiety engaged in a π-stacking interaction with Arg55 of cyclophilin A (Cyp A), and the m-Tyr residue was displaced into solvent. This observation allowed further simplifications of the scaffold to generate new lead compounds in the search for orally bioavailable cyclophilin inhibitors.


Asunto(s)
Ciclofilinas/antagonistas & inhibidores , Células Cultivadas , Cromatografía Liquida , Cristalografía por Rayos X , Descubrimiento de Drogas , Humanos , Enlace de Hidrógeno , Lactonas/química , Lactonas/farmacología , Espectroscopía de Protones por Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Termodinámica
11.
PLoS One ; 9(1): e84808, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24416288

RESUMEN

Elucidation of the mechanism of action of the HCV NS5B polymerase thumb site II inhibitors has presented a challenge. Current opinion holds that these allosteric inhibitors stabilize the closed, inactive enzyme conformation, but how this inhibition is accomplished mechanistically is not well understood. Here, using a panel of NS5B proteins with mutations in key regulatory motifs of NS5B--the C-terminal tail and ß-loop--in conjunction with a diverse set of NS5B allosteric inhibitors, we show that thumb site II inhibitors possess a distinct mechanism of action. A combination of enzyme activity studies and direct binding assays reveals that these inhibitors require both regulatory elements to maintain the polymerase inhibitory activity. Removal of either element has little impact on the binding affinity of thumb site II inhibitors, but significantly reduces their potency. NS5B in complex with a thumb site II inhibitor displays a characteristic melting profile that suggests stabilization not only of the thumb domain but also the whole polymerase. Successive truncations of the C-terminal tail and/or removal of the ß-loop lead to progressive destabilization of the protein. Furthermore, the thermal unfolding transitions characteristic for thumb site II inhibitor-NS5B complex are absent in the inhibitor-bound constructs in which interactions between C-terminal tail and ß-loop are abolished, pointing to the pivotal role of both regulatory elements in communication between domains. Taken together, a comprehensive picture of inhibition by compounds binding to thumb site II emerges: inhibitor binding provides stabilization of the entire polymerase in an inactive, closed conformation, propagated via coupled interactions between the C-terminal tail and ß-loop.


Asunto(s)
Sitio Alostérico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hepacivirus/enzimología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/química , Secuencias de Aminoácidos , Dominio Catalítico , Estabilidad de Enzimas , Furanos/farmacología , Modelos Moleculares , Eliminación de Secuencia , Tiofenos/farmacología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
12.
PLoS One ; 7(6): e39163, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22720059

RESUMEN

Tegobuvir (TGV) is a novel non-nucleoside inhibitor (NNI) of HCV RNA replication with demonstrated antiviral activity in patients with genotype 1 chronic HCV infection. The mechanism of action of TGV has not been clearly defined despite the identification of resistance mutations mapping to the NS5B polymerase region. TGV does not inhibit NS5B enzymatic activity in biochemical assays in vitro, suggesting a more complex antiviral mechanism with cellular components. Here, we demonstrate that TGV exerts anti-HCV activity utilizing a unique chemical activation and subsequent direct interaction with the NS5B protein. Treatment of HCV subgenomic replicon cells with TGV results in a modified form of NS5B with a distinctly altered mobility on a SDS-PAGE gel. Further analysis reveals that the aberrantly migrating NS5B species contains the inhibitor molecule. Formation of this complex does not require the presence of any other HCV proteins. The intensity of the aberrantly migrating NS5B species is strongly dependent on cellular glutathione levels as well as CYP 1A activity. Furthermore analysis of NS5B protein purified from a heterologous expression system treated with TGV by mass spectrometry suggests that TGV undergoes a CYP- mediated intracellular activation step and the resulting metabolite, after forming a glutathione conjugate, directly and specifically interacts with NS5B. Taken together, these data demonstrate that upon metabolic activation TGV is a specific, covalent inhibitor of the HCV NS5B polymerase and is mechanistically distinct from other classes of the non-nucleoside inhibitors (NNI) of the viral polymerase.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Purinas/farmacología , Piridazinas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Humanos , Espectrometría de Masas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
ACS Med Chem Lett ; 3(7): 565-9, 2012 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-24900511

RESUMEN

A series of imidazo[1,2-a]pyridines which directly bind to HCV Non-Structural Protein 4B (NS4B) is described. This series demonstrates potent in vitro inhibition of HCV replication (EC50 < 10 nM), direct binding to purified NS4B protein (IC50 < 20 nM), and an HCV resistance pattern associated with NS4B (H94N/R, V105L/M, F98L) that are unique among reported HCV clinical assets, suggestive of the potential for additive or synergistic combination with other small molecule inhibitors of HCV replication.

14.
Antimicrob Agents Chemother ; 55(9): 4196-203, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21746939

RESUMEN

GS-9190 (Tegobuvir) is a novel imidazopyridine inhibitor of hepatitis C virus (HCV) RNA replication in vitro and has demonstrated potent antiviral activity in patients chronically infected with genotype 1 (GT1) HCV. GS-9190 exhibits reduced activity against GT2a (JFH1) subgenomic replicons and GT2a (J6/JFH1) infectious virus, suggesting that the compound's mechanism of action involves a genotype-specific viral component. To further investigate the GS-9190 mechanism of action, we utilized the susceptibility differences between GT1b and GT2a by constructing a series of replicon chimeras where combinations of 1b and 2a nonstructural proteins were encoded within the same replicon. The antiviral activities of GS-9190 against the chimeric replicons were reduced to levels comparable to that of the wild-type GT2a replicon in chimeras expressing GT2a NS5B. GT1b replicons in which the ß-hairpin region (amino acids 435 to 455) was replaced by the corresponding sequence of GT2a were markedly less susceptible to GS-9190, indicating the importance of the thumb subdomain of the polymerase in this effect. Resistance selection in GT1b replicon cells identified several mutations in NS5B (C316Y, Y448H, Y452H, and C445F) that contributed to the drug resistance phenotype. Reintroduction of these mutations into wild-type replicons conferred resistance to GS-9190, with the number of NS5B mutations correlating with the degree of resistance. Analysis of GS-9190 cross-resistance against previously reported NS5B drug-selected mutations showed that the resistance pattern of GS-9190 is different from other nonnucleoside inhibitors. Collectively, these data demonstrate that GS-9190 represents a novel class of nonnucleoside polymerase inhibitors that interact with NS5B likely through involvement of the ß-hairpin in the thumb subdomain.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Purinas/farmacología , Piridazinas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Antivirales/química , Línea Celular , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Mutación , Plásmidos/genética , Purinas/química , Piridazinas/química
15.
Antiviral Res ; 87(1): 1-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20363257

RESUMEN

Hepatitis C Virus (HCV) is a leading cause of liver disease and represents a significant public health challenge. Treatments for this disease are inadequate and improved antiviral therapies are necessary. Several such antivirals are in development, most of which target the well-characterized NS3 protease or the NS5B polymerase. In contrast, the nonstructural 4B (NS4B) protein, though essential for HCV RNA replication, has been the subject of few pharmacological studies. One of the functions ascribed to this protein is the ability to form intracellular membrane-associated foci (MAF), which are believed to be related to the sites of viral replication. Here, we report the identification of a small molecule that inhibits HCV replication and disrupts the organization of these MAF. Genetic analysis links the compound's mode of action to the NS4B gene product, and transient transfections of NS4B-GFP demonstrate that treatment with this compound can lead to the formation of novel elongated assemblies of NS4B. Furthermore, an in vitro dynamic light scattering assay provides evidence that the second amphipathic helix of NS4B may be the target of the drug. Our results demonstrate that this molecule represents a new potential class of HCV inhibitors and also provides us with a useful tool for studying the HCV life cycle.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos , Fusión Artificial Génica , Células Cultivadas , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hepatocitos/virología , Humanos , Modelos Biológicos , Modelos Moleculares , Estructura Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
16.
Recent Pat Antiinfect Drug Discov ; 3(2): 77-92, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18673121

RESUMEN

The hepatitis C virus (HCV) non-structural 5A (NS5A) protein is essential for viral RNA replication and may play a role in subverting host intracellular signaling pathways. Although no intrinsic enzymatic activity has been ascribed to NS5A, this proline-rich hydrophilic phosphoprotein is likely to exert its functions by interacting with viral and cellular factors. Recent studies using the HCV replicon cell culture system as a model for HCV RNA replication as well as for high-throughput screening of pharmacological inhibitors have revealed blockade of NS5A as a promising therapeutic strategy for the treatment of HCV. This review will summarize our progress in understanding the role of NS5A in HCV RNA replication and will introduce the most recent patents on inhibitors of NS5A.


Asunto(s)
Antivirales/uso terapéutico , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/química , Diseño de Fármacos , Farmacorresistencia Viral , Hepacivirus/crecimiento & desarrollo , Hepacivirus/metabolismo , Hepatitis C/virología , Humanos , Modelos Moleculares , Estructura Molecular , Mutación , Patentes como Asunto , Conformación Proteica , ARN Viral/biosíntesis , Relación Estructura-Actividad , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
17.
J Med Chem ; 45(4): 805-17, 2002 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-11831893

RESUMEN

A new series of short pyrrole tetraamides are described whose submicromolar DNA binding affinity is an essential component for their strong antibacterial activity. This class of compounds is related to the linked bis-netropsins and bis-distamycins, but here, only one amino-pyrrole-carboxamide unit and an amidine tail is connected to either side of a central dicarboxylic acid linker. The highest degree of DNA binding, measured by compound-induced changes in UV melting temperatures of an AT-rich DNA oligomer, was observed for flat, aromatic linkers with no inherent bent, i.e., terephthalic acid or 1,4-pyridine-dicarboxylic acid. However, the antibacterial activity is critically linked to the size of the N-alkyl substiutent of the pyrrole unit. None of the tetraamides with the commonly used methyl-pyrrole showed antibacterial activity. Isoamyl- or cyclopropylmethylene-substituted dipyrrole derivatives have the minimum inhibitory concentrations in the submicromolar range. In vitro toxicity against human T-cells was studied for all compounds. The degree to which compounds inhibited cell growth was neither directly correlated to DNA binding affinity nor directly correlated to antibacterial activity but seemed to depend strongly on the nature of the N-alkyl pyrrole substituents.


Asunto(s)
Amidas/síntesis química , Antibacterianos/síntesis química , ADN/química , Enterococcus/efectos de los fármacos , Pirroles/síntesis química , Staphylococcus aureus/efectos de los fármacos , Amidas/química , Amidas/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Humanos , Resistencia a la Meticilina , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Pirroles/química , Pirroles/farmacología , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Células Tumorales Cultivadas , Resistencia a la Vancomicina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...