Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Cent Sci ; 10(1): 87-103, 2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38292603

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with no cure, and current treatment options are very limited. Previously, we performed a high-throughput screen to identify small molecules that inhibit protein aggregation caused by a mutation in the gene that encodes superoxide dismutase 1 (SOD1), which is responsible for about 25% of familial ALS. This resulted in three hit series of compounds that were optimized over several years to give three compounds that were highly active in a mutant SOD1 ALS model. Here we identify the target of two of the active compounds (6 and 7) with the use of photoaffinity labeling, chemical biology reporters, affinity purification, proteomic analysis, and fluorescent/cellular thermal shift assays. Evidence is provided to demonstrate that these two pyrazolone compounds directly interact with 14-3-3-E and 14-3-3-Q isoforms, which have chaperone activity and are known to interact with mutant SOD1G93A aggregates and become insoluble in the subcellular JUNQ compartment, leading to apoptosis. Because protein aggregation is the hallmark of all neurodegenerative diseases, knowledge of the target compounds that inhibit protein aggregation allows for the design of more effective molecules for the treatment of ALS and possibly other neurodegenerative diseases.

2.
J Am Chem Soc ; 143(21): 8193-8207, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34014654

RESUMEN

Human ornithine aminotransferase (hOAT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that was recently found to play an important role in the metabolic reprogramming of hepatocellular carcinoma (HCC) via the proline and glutamine metabolic pathways. The selective inhibition of hOAT by compound 10 exhibited potent in vivo antitumor activity. Inspired by the discovery of the aminotransferase inactivator (1S,3S)-3-amino-4-(difluoromethylene)cyclopentane-1-carboxylic acid (5), we rationally designed, synthesized, and evaluated a series of six-membered-ring analogs. Among them, 14 was identified as a new selective hOAT inactivator, which demonstrated a potency 22× greater than that of 10. Three different types of protein mass spectrometry approaches and two crystallographic approaches were employed to identify the structure of hOAT-14 and the formation of a remarkable final adduct (32') in the active site. These spectral studies reveal an enzyme complex heretofore not observed in a PLP-dependent enzyme, which has covalent bonds to two nearby residues. Crystal soaking experiments and molecular dynamics simulations were carried out to identify the structure of the active-site intermediate 27' and elucidate the order of the two covalent bonds that formed, leading to 32'. The initial covalent reaction of the activated warhead occurs with *Thr322 from the second subunit, followed by a subsequent nucleophilic attack by the catalytic residue Lys292. The turnover mechanism of 14 by hOAT was supported by a mass spectrometric analysis of metabolites and fluoride ion release experiments. This novel mechanism for hOAT with 14 will contribute to the further rational design of selective inactivators and an understanding of potential inactivation mechanisms by aminotransferases.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Ornitina-Oxo-Ácido Transaminasa/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Cinética , Espectrometría de Masas , Modelos Moleculares , Estructura Molecular , Ornitina-Oxo-Ácido Transaminasa/metabolismo
3.
ACS Chem Biol ; 16(4): 615-630, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33735567

RESUMEN

((S)-3-Amino-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (OV329) is a recently discovered inactivator of γ-aminobutyric acid aminotransferase (GABA-AT), which has 10 times better inactivation efficiency than its predecessor, CPP-115, despite the only structural difference being an endocyclic double bond in OV329. Both compounds are mechanism-based enzyme inactivators (MBEIs), which inactivate GABA-AT by a similar mechanism. Here, a combination of a variety of computational chemistry tools and experimental methods, including quantum mechanical (QM) calculations, molecular dynamic simulations, progress curve analysis, and deuterium kinetic isotope effect (KIE) experiments, are utilized to comprehensively study the mechanism of inactivation of GABA-AT by CPP-115 and OV329 and account for their experimentally obtained global kinetic parameters kinact and KI. Our first key finding is that the rate-limiting step of the inactivation mechanism is the deprotonation step, and according to QM calculations and the KIE experiments, kinact accurately represents the enhancement of the rate-limiting step for the given mechanism. Second, the present study shows that the widely used simple QM models do not accurately represent the geometric criteria that are present in the enzyme for the deprotonation step. In contrast, QM cluster models successfully represent both the ground state destabilization and the transition state stabilization, as revealed by natural bond orbital analysis. Furthermore, the globally derived KI values for both of the inactivators represent the inhibitor constants for the initial binding complexes (Kd) and indicate the inactivator competition with the substrate according to progress curve analysis and the observed binding isotope effect. The configurational entropy loss accounts for the difference in KI values between the inactivators. The approach we describe in this work can be employed to determine the validity of globally derived parameters in the process of MBEI optimization for given inactivation mechanisms.


Asunto(s)
4-Aminobutirato Transaminasa/metabolismo , Prolina/análogos & derivados , 4-Aminobutirato Transaminasa/antagonistas & inhibidores , Catálisis , Cinética , Simulación de Dinámica Molecular , Prolina/farmacología , Teoría Cuántica , Reproducibilidad de los Resultados
4.
ACS Med Chem Lett ; 11(10): 1949-1955, 2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33062178

RESUMEN

Aminotransferases are pyridoxal 5'-phosphate-dependent enzymes that catalyze reversible transamination reactions between an amino acid and an α-keto acid, playing a critical role in cellular nitrogen metabolism. It is evident that γ-aminobutyric acid aminotransferase (GABA-AT), which balances the levels of inhibitory and excitatory neurotransmitters, has emerged as a promising therapeutic target for epilepsy and cocaine addiction based on mechanism-based inactivators (MBIs). In this work, we established an integrated approach using computational simulation, organic synthesis, biochemical evaluation, and mass spectrometry to facilitate our design and mechanistic studies of MBIs, which led to the identification of a new cyclopentene-based analogue (6a), 25-times more efficient as an inactivator of GABA-AT compared to the parent compound (1R,3S,4S)-3-amino-4-fluorocyclopentane carboxylic acid (FCP, 4).

5.
J Am Chem Soc ; 142(10): 4892-4903, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32114761

RESUMEN

Human ornithine aminotransferase (hOAT), a pyridoxal 5'-phosphate-dependent enzyme, plays a critical role in the progression of hepatocellular carcinoma (HCC). Pharmacological selective inhibition of hOAT has been shown to be a potential therapeutic approach for HCC. Inspired by the discovery of the nonselective aminotransferase inactivator (1R,3S,4S)-3-amino-4-fluoro cyclopentane-1-carboxylic acid (1), in this work, we rationally designed, synthesized, and evaluated a novel series of fluorine-substituted cyclohexene analogues, thereby identifying 8 and 9 as novel selective hOAT time-dependent inhibitors. Intact protein mass spectrometry and protein crystallography demonstrated 8 and 9 as covalent inhibitors of hOAT, which exhibit two distinct inactivation mechanisms resulting from the difference of a single fluorine atom. Interestingly, they share a similar turnover mechanism, according to the mass spectrometry-based analysis of metabolites and fluoride ion release experiments. Molecular dynamics (MD) simulations and electrostatic potential (ESP) charge calculations were conducted, which elucidated the significant influence of the one-fluorine difference on the corresponding intermediates, leading to two totally different inactivation pathways. The novel addition-aromatization inactivation mechanism for 9 contributes to its significantly enhanced potency, along with excellent selectivity over other aminotransferases.


Asunto(s)
Ácidos Ciclohexanocarboxílicos/química , Ciclohexilaminas/química , Inhibidores Enzimáticos/química , Hidrocarburos Fluorados/química , Ornitina-Oxo-Ácido Transaminasa/antagonistas & inhibidores , Ácidos Ciclohexanocarboxílicos/síntesis química , Ácidos Ciclohexanocarboxílicos/metabolismo , Ciclohexilaminas/síntesis química , Ciclohexilaminas/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/metabolismo , Humanos , Hidrocarburos Fluorados/síntesis química , Hidrocarburos Fluorados/metabolismo , Modelos Químicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Ornitina-Oxo-Ácido Transaminasa/química , Ornitina-Oxo-Ácido Transaminasa/metabolismo , Unión Proteica , Fosfato de Piridoxal/química , Ácido gamma-Aminobutírico/análogos & derivados
6.
Proteins ; 87(7): 579-587, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30883881

RESUMEN

Human noroviruses are the primary cause of outbreaks of acute gastroenteritis worldwide. The problem is further compounded by the current lack of norovirus-specific antivirals or vaccines. Noroviruses have a single-stranded, positive sense 7 to 8 kb RNA genome which encodes a polyprotein precursor that is processed by a virus-encoded 3C-like cysteine protease (NV 3CLpro) to generate at least six mature nonstructural proteins. Processing of the polyprotein is essential for virus replication, consequently, NV 3CLpro has emerged as an attractive target for the discovery of norovirus therapeutics and prophylactics. We have recently described the structure-based design of macrocyclic transition state inhibitors of NV 3CLpro. In order to gain insight and understanding into the interaction of macrocyclic inhibitors with the enzyme, as well as probe the effect of ring size on pharmacological activity and cellular permeability, additional macrocyclic inhibitors were synthesized and high resolution cocrystal structures determined. The results of our studies tentatively suggest that the macrocyclic scaffold may hamper optimal binding to the active site by impeding concerted cross-talk between the S2 and S4 subsites.


Asunto(s)
Proteasas de Cisteína/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Compuestos Macrocíclicos/farmacología , Norovirus/enzimología , Animales , Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/virología , Dominio Catalítico/efectos de los fármacos , Línea Celular , Cristalografía por Rayos X , Proteasas de Cisteína/química , Inhibidores de Cisteína Proteinasa/química , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/virología , Humanos , Compuestos Macrocíclicos/química , Ratones , Modelos Moleculares , Norovirus/química , Norovirus/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Células RAW 264.7
7.
J Am Chem Soc ; 140(6): 2151-2164, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29381352

RESUMEN

γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system. Inhibition of GABA aminotransferase (GABA-AT), a pyridoxal 5'-phosphate (PLP)-dependent enzyme that degrades GABA, has been established as a possible strategy for the treatment of substance abuse. The raised GABA levels that occur as a consequence of this inhibition have been found to antagonize the rapid release of dopamine in the ventral striatum (nucleus accumbens) that follows an acute challenge by an addictive substance. In addition, increased GABA levels are also known to elicit an anticonvulsant effect in patients with epilepsy. We previously designed the mechanism-based inactivator (1S,3S)-3-amino-4-difluoromethylenyl-1-cyclopentanoic acid (2), now called CPP-115, that is 186 times more efficient in inactivating GABA-AT than vigabatrin, the only FDA-approved drug that is an inactivator of GABA-AT. CPP-115 was found to have high therapeutic potential for the treatment of cocaine addiction and for a variety of epilepsies, has successfully completed a Phase I safety clinical trial, and was found to be effective in the treatment of infantile spasms (West syndrome). Herein we report the design, using molecular dynamics simulations, synthesis, and biological evaluation of a new mechanism-based inactivator, (S)-3-amino-4-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic acid (5), which was found to be almost 10 times more efficient as an inactivator of GABA-AT than CPP-115. We also present the unexpected crystal structure of 5 bound to GABA-AT, as well as computational analyses used to assist the structure elucidation process. Furthermore, 5 was found to have favorable pharmacokinetic properties and low off-target activities. In vivo studies in freely moving rats showed that 5 was dramatically superior to CPP-115 in suppressing the release of dopamine in the corpus striatum, which occurs subsequent to either an acute cocaine or nicotine challenge. Compound 5 also attenuated increased metabolic demands (neuronal glucose metabolism) in the hippocampus, a brain region that encodes spatial information concerning the environment in which an animal receives a reinforcing or aversive drug. This multidisciplinary computational design to preclinical efficacy approach should be applicable to the design and improvement of mechanism-based inhibitors of other enzymes whose crystal structures and inactivation mechanisms are known.


Asunto(s)
4-Aminobutirato Transaminasa/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Prolina/análogos & derivados , 4-Aminobutirato Transaminasa/química , 4-Aminobutirato Transaminasa/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Dopamina/metabolismo , Antagonistas de Dopamina/química , Antagonistas de Dopamina/farmacocinética , Antagonistas de Dopamina/farmacología , Inhibidores Enzimáticos/farmacocinética , Glucosa/metabolismo , Humanos , Masculino , Modelos Moleculares , Prolina/química , Prolina/farmacocinética , Prolina/farmacología , Ratas , Ratas Sprague-Dawley , Ácido gamma-Aminobutírico/metabolismo
8.
Eur J Med Chem ; 126: 502-516, 2017 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-27914364

RESUMEN

Human noroviruses are the primary cause of epidemic and sporadic acute gastroenteritis. The worldwide high morbidity and mortality associated with norovirus infections, particularly among the elderly, immunocompromised patients and children, constitute a serious public health concern. There are currently no approved human vaccines or norovirus-specific small-molecule therapeutics or prophylactics. Norovirus 3CL protease has recently emerged as a potential therapeutic target for the development of anti-norovirus agents. We hypothesized that the S4 subsite of the enzyme may provide an effective means of designing potent and cell permeable inhibitors of the enzyme. We report herein the structure-guided exploration and exploitation of the S4 subsite of norovirus 3CL protease in the design and synthesis of effective inhibitors of the protease.


Asunto(s)
Diseño de Fármacos , Norovirus/enzimología , Péptido Hidrolasas/química , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Línea Celular , Humanos , Modelos Moleculares , Norovirus/efectos de los fármacos , Norovirus/fisiología , Permeabilidad , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/toxicidad , Conformación Proteica , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
9.
Eur J Med Chem ; 119: 300-18, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27235842

RESUMEN

Outbreaks of acute gastroenteritis caused by noroviruses constitute a public health concern worldwide. To date, there are no approved drugs or vaccines for the management and prophylaxis of norovirus infections. A potentially effective strategy for the development of norovirus therapeutics entails the discovery of inhibitors of norovirus 3CL protease, an enzyme essential for noroviral replication. We describe herein the structure-based design of the first class of permeable, triazole-based macrocyclic inhibitors of norovirus 3C-like protease, as well as pertinent X-ray crystallographic, biochemical, spectroscopic, and antiviral studies.


Asunto(s)
Diseño de Fármacos , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Norovirus/efectos de los fármacos , Péptido Hidrolasas/metabolismo , Triazoles/química , Técnicas de Química Sintética , Compuestos Macrocíclicos/metabolismo , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Péptido Hidrolasas/química , Permeabilidad , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Conformación Proteica
10.
Expert Opin Ther Pat ; 26(3): 297-308, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26881878

RESUMEN

INTRODUCTION: Human noroviruses are the primary causative agents of acute gastroenteritis and are a pressing public health burden worldwide. There are currently no vaccines or small molecule therapeutics available for the treatment or prophylaxis of norovirus infections. An improved understanding of norovirus biology, as well as the pathogenic mechanisms underlying the disease, has provided the impetus for a range of intense exploratory drug discovery efforts targeting viral and host factors. AREAS COVERED: An overview of norovirus inhibitors disclosed in the patent literature (2010-present) and Clinicaltrials.gov is presented. The review is further enriched and supplemented by recent literature reports. EXPERT OPINION: Seminal discoveries made in recent years, including a better understanding of the pathobiology and life cycle of norovirus, the identification and targeting of multiple viral and host factors, the advent of a replicon system and a small animal model for the preclinical evaluation of lead compounds, and the availability of high resolution X-ray crystal structures that can be utilized in structure-based drug design and lead optimization campaigns, collectively suggest that a small molecule therapeutic and prophylactic for norovirus infection is likely to emerge in the not too distant future.


Asunto(s)
Antivirales/farmacología , Infecciones por Caliciviridae/tratamiento farmacológico , Norovirus/efectos de los fármacos , Animales , Infecciones por Caliciviridae/virología , Diseño de Fármacos , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/virología , Humanos , Patentes como Asunto
11.
J Med Chem ; 59(5): 1899-913, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26823007

RESUMEN

Human noroviruses are the primary causative agents of acute gastroenteritis and a pressing public health burden worldwide. There are currently no vaccines or small molecule therapeutics available for the treatment or prophylaxis of norovirus infections. Norovirus 3CL protease plays a vital role in viral replication by generating structural and nonstructural proteins via the cleavage of the viral polyprotein. Thus, molecules that inhibit the viral protease may have potential therapeutic value. We describe herein the structure-based design, synthesis, and in vitro and cell-based evaluation of the first class of oxadiazole-based, permeable macrocyclic inhibitors of norovirus 3CL protease.


Asunto(s)
Antivirales/farmacología , Permeabilidad de la Membrana Celular , Compuestos Macrocíclicos/farmacología , Norovirus/efectos de los fármacos , Norovirus/enzimología , Oxadiazoles/farmacología , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/farmacología , Animales , Antivirales/síntesis química , Antivirales/química , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Humanos , Compuestos Macrocíclicos/síntesis química , Compuestos Macrocíclicos/química , Ratones , Modelos Moleculares , Estructura Molecular , Oxadiazoles/síntesis química , Oxadiazoles/química , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Relación Estructura-Actividad
12.
Antiviral Res ; 125: 84-91, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26658373

RESUMEN

Enterovirus D68 (EV-D68) is an emerging pathogen responsible for mild to severe respiratory infections that occur mostly in infants, children and teenagers. EV-D68, one of more than 100 non-polio enteroviruses, is acid-labile and biologically similar to human rhinoviruses (HRV) (originally classified as HRV87). However, there is no approved preventive or therapeutic measure against EV-D68, HRV, or other enteroviruses. In this study, we evaluated the antiviral activity of series of dipeptidyl compounds against EV-D68 and HRV strains, and demonstrated that several peptidyl aldehyde and α-ketoamide peptidyl compounds are potent inhibitors of EV-D68 and HRV strains with high in-vitro therapeutic indices (>1000). One of the α-ketoamide compounds is shown to have favorable pharmacokinetics profiles, including a favorable oral bioavailability in rats. Recent successful development of α-ketoamide protease inhibitors against hepatitis C virus suggests these compounds may have a high potential for further optimization and development against emerging EV-D68, as well as HRV.


Asunto(s)
Aldehídos/farmacología , Amidas/farmacología , Dipéptidos/farmacología , Enterovirus Humano D/efectos de los fármacos , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Picornaviridae/tratamiento farmacológico , Rhinovirus/efectos de los fármacos , Aldehídos/síntesis química , Aldehídos/farmacocinética , Amidas/síntesis química , Amidas/farmacocinética , Animales , Antivirales/farmacología , Dipéptidos/síntesis química , Dipéptidos/farmacocinética , Quimioterapia Combinada , Infecciones por Enterovirus/virología , Femenino , Cobayas , Células HeLa , Humanos , Modelos Moleculares , Infecciones por Picornaviridae/virología , Ratas Sprague-Dawley , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/virología
13.
J Med Chem ; 58(7): 3144-55, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25761614

RESUMEN

Norovirus infection constitutes the primary cause of acute viral gastroenteritis. There are currently no vaccines or norovirus-specific antiviral therapeutics available for the management of norovirus infection. Norovirus 3C-like protease is essential for viral replication, consequently, inhibition of this enzyme is a fruitful avenue of investigation that may lead to the emergence of antinorovirus therapeutics. We describe herein the optimization of dipeptidyl inhibitors of norovirus 3C-like protease using iterative SAR, X-ray crystallographic, and enzyme and cell-based studies. We also demonstrate herein in vivo efficacy of an inhibitor using the murine model of norovirus infection.


Asunto(s)
Norovirus/enzimología , Péptido Hidrolasas/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Proteínas Virales/química , Animales , Antivirales/química , Antivirales/farmacología , Línea Celular/efectos de los fármacos , Técnicas de Química Sintética , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/química , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Femenino , Macrófagos/efectos de los fármacos , Macrófagos/virología , Ratones Endogámicos BALB C , Modelos Moleculares , Norovirus/efectos de los fármacos , Norovirus/patogenicidad , Péptido Hidrolasas/metabolismo , Conformación Proteica , Relación Estructura-Actividad , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo
14.
Bioorg Med Chem Lett ; 23(13): 3709-12, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23727045

RESUMEN

The design, synthesis, and in vitro evaluation of the first macrocyclic inhibitor of 3C and 3C-like proteases of picornavirus, norovirus, and coronavirus are reported. The in vitro inhibitory activity (50% effective concentration) of the macrocyclic inhibitor toward enterovirus 3C protease (CVB3 Nancy strain), and coronavirus (SARS-CoV) and norovirus 3C-like proteases, was determined to be 1.8, 15.5 and 5.1 µM, respectively.


Asunto(s)
Coronavirus/enzimología , Compuestos Macrocíclicos/farmacología , Norovirus/enzimología , Péptido Hidrolasas/metabolismo , Picornaviridae/enzimología , Inhibidores de Proteasas/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Compuestos Macrocíclicos/síntesis química , Compuestos Macrocíclicos/química , Modelos Moleculares , Conformación Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , Relación Estructura-Actividad
15.
Bioorg Med Chem Lett ; 22(14): 4820-6, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22698498

RESUMEN

A series of structurally-diverse α-ketoamides and α-ketoheterocycles was synthesized and subsequently investigated for inhibitory activity against norovirus 3CL protease in vitro, as well as anti-norovirus activity in a cell-based replicon system. The synthesized compounds were found to inhibit norovirus 3CL protease in vitro and to also exhibit potent anti-norovirus activity in a cell-based replicon system.


Asunto(s)
Amidas/química , Cisteína Endopeptidasas/química , Compuestos Heterocíclicos/química , Norovirus/enzimología , Péptidos/química , Inhibidores de Proteasas/química , Amidas/farmacología , Cisteína Endopeptidasas/farmacología , Compuestos Heterocíclicos/farmacología , Modelos Moleculares , Estructura Molecular , Norovirus/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...