Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Med Chem ; 64(19): 14465-14476, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1894373

RESUMEN

In this work, a series of novel substituted polycyclic pyridone derivatives were designed and synthesized as potent anti-influenza agents. The cytopathic effect (CPE) assay and cytotoxicity assay indicated that all of the compounds possessed potent anti-influenza virus activity and relatively low cytotoxicity; some of them inhibited the replication of influenza A virus (IAV) at picomolar concentrations. Further studies revealed that, at a concentration of 3 nM, three compounds (10a, 10d, and 10g) could significantly reduce the M2 RNA amounts and M2 protein expression of IAV and inhibit the activity of RNA-dependent RNA polymerase (RdRp). Among them, (R)-12-(5H-dibenzo[a,d][7]annulen-5-yl)-7-hydroxy-3,4,12,12a-tetrahydro-1H-[1,4]oxazino[3,4-c]pyrido[2,1-f][1,2,4]triazine-6,8-dione (10a) was found to be a promising anti-influenza drug candidate with good human liver microsomal stability, as well as with better selectivity index and oral bioavailability than Baloxavir.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Dibenzotiepinas/química , Virus de la Influenza A/efectos de los fármacos , Morfolinas/química , Piridonas/síntesis química , Piridonas/farmacología , Triazinas/química , Animales , Supervivencia Celular/efectos de los fármacos , Efecto Citopatogénico Viral/efectos de los fármacos , Perros , Humanos , Células de Riñón Canino Madin Darby , Masculino , Piridonas/química , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
Structure ; 29(8): 823-833.e5, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1275725

RESUMEN

There is a clinical need for direct-acting antivirals targeting SARS-CoV-2, the coronavirus responsible for the COVID-19 pandemic, to complement current therapeutic strategies. The main protease (Mpro) is an attractive target for antiviral therapy. However, the vast majority of protease inhibitors described thus far are peptidomimetic and bind to the active-site cysteine via a covalent adduct, which is generally pharmacokinetically unfavorable. We have reported the optimization of an existing FDA-approved chemical scaffold, perampanel, to bind to and inhibit Mpro noncovalently with IC50s in the low-nanomolar range and EC50s in the low-micromolar range. Here, we present nine crystal structures of Mpro bound to a series of perampanel analogs, providing detailed structural insights into their mechanism of action and structure-activity relationship. These insights further reveal strategies for pursuing rational inhibitor design efforts in the context of considerable active-site flexibility and potential resistance mechanisms.


Asunto(s)
Antivirales/química , Proteasas 3C de Coronavirus/química , Inhibidores de Proteasas/química , Piridonas/química , SARS-CoV-2/enzimología , Antivirales/farmacología , COVID-19/virología , Dominio Catalítico , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/metabolismo , Diseño de Fármacos , Simulación de Dinámica Molecular , Estructura Molecular , Nitrilos , Inhibidores de Proteasas/farmacología , Conformación Proteica , Multimerización de Proteína , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Tratamiento Farmacológico de COVID-19
3.
Comb Chem High Throughput Screen ; 24(3): 441-454, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1102440

RESUMEN

BACKGROUND: Coronavirus Disease 2019 (COVID-19) pandemic continues to threaten patients, societies and healthcare systems around the world. There is an urgent need to search for possible medications. OBJECTIVE: This article intends to use virtual screening and molecular docking methods to find potential inhibitors from existing drugs that can respond to COVID-19. METHODS: To take part in the current research investigation and to define a potential target drug that may protect the world from the pandemic of corona disease, a virtual screening study of 129 approved drugs was carried out which showed that their metabolic characteristics, dosages used, potential efficacy and side effects are clear as they have been approved for treating existing infections. Especially 12 drugs against chronic hepatitis B virus, 37 against chronic hepatitis C virus, 37 against human immunodeficiency virus, 14 anti-herpesvirus, 11 anti-influenza, and 18 other drugs currently on the market were considered for this study. These drugs were then evaluated using virtual screening and molecular docking studies on the active site of the (SARS-CoV-2) main protease (6lu7). Once the efficacy of the drug is determined, it can be approved for its in vitro and in vivo activity against the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), which can be beneficial for the rapid clinical treatment of patients. These drugs were considered potentially effective against SARS-CoV-2 and those with high molecular docking scores were proposed as novel candidates for repurposing. The N3 inhibitor cocrystallized with protease (6lu7) and the anti-HIV protease inhibitor Lopinavir were used as standards for comparison. RESULTS: The results suggest the effectiveness of Beclabuvir, Nilotinib, Tirilazad, Trametinib and Glecaprevir as potent drugs against SARS-CoV-2 since they tightly bind to its main protease. CONCLUSION: These promising drugs can inhibit the replication of the virus; hence, the repurposing of these compounds is suggested for the treatment of COVID-19. No toxicity measurements are required for these drugs since they were previously tested prior to their approval by the FDA. However, the assessment of these potential inhibitors as clinical drugs requires further in vivo tests of these drugs.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Proteasas 3C de Coronavirus/metabolismo , Evaluación Preclínica de Medicamentos/métodos , SARS-CoV-2/efectos de los fármacos , Antivirales/metabolismo , Sitios de Unión , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Reposicionamiento de Medicamentos , Hepacivirus/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Lopinavir/química , Lopinavir/farmacología , Simulación del Acoplamiento Molecular , Piridonas/química , Piridonas/farmacología , Pirimidinonas/química , Pirimidinonas/farmacología
4.
Chem Biol Interact ; 335: 109348, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: covidwho-956088

RESUMEN

The disease, COVID-19, is caused by the severe acute respiratory coronavirus 2 (SARS-CoV-2) for which there is currently no treatment. The SARS-CoV-2 main protease (Mpro) is an important enzyme for viral replication. Small molecules that inhibit this protease could lead to an effective COVID-19 treatment. The 2-pyridone scaffold was previously identified as a possible key pharmacophore to inhibit SARS-CoV-2 Mpro. A search for natural, antimicrobial products with the 2-pyridone moiety was undertaken herein, and their calculated potency as inhibitors of SARS-CoV-2 Mpro was investigated. Thirty-three natural products containing the 2-pyridone scaffold were identified from the literature. An in silico methodology using AutoDock was employed to predict the binding energies and inhibition constants (Ki values) for each 2-pyridone-containing compound with SARS-CoV-2 Mpro. This consisted of molecular optimization of the 2-pyridone compound, docking of the compound with a crystal structure of SARS-CoV-2 Mpro, and evaluation of the predicted interactions and ligand-enzyme conformations. All compounds investigated bound to the active site of SARS-CoV-2 Mpro, close to the catalytic dyad (His-41 and Cys-145). Thirteen molecules had predicted Ki values <1 µM. Glu-166 formed a key hydrogen bond in the majority of the predicted complexes, while Met-165 had some involvement in the complex binding as a close contact to the ligand. Prominent 2-pyridone compounds were further evaluated for their ADMET properties. This work has identified 2-pyridone natural products with calculated potent inhibitory activity against SARS-CoV-2 Mpro and with desirable drug-like properties, which may lead to the rapid discovery of a treatment for COVID-19.


Asunto(s)
Antivirales/metabolismo , Productos Biológicos/metabolismo , Proteasas 3C de Coronavirus/metabolismo , Inhibidores de Cisteína Proteinasa/metabolismo , Piridonas/metabolismo , SARS-CoV-2/enzimología , Antivirales/química , Antivirales/farmacocinética , Productos Biológicos/química , Productos Biológicos/farmacocinética , Células CACO-2 , Dominio Catalítico , Proteasas 3C de Coronavirus/química , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacocinética , Humanos , Enlace de Hidrógeno , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Piridonas/química , Piridonas/farmacocinética
5.
Science ; 368(6489): 409-412, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: covidwho-164984

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is a global health emergency. An attractive drug target among coronaviruses is the main protease (Mpro, also called 3CLpro) because of its essential role in processing the polyproteins that are translated from the viral RNA. We report the x-ray structures of the unliganded SARS-CoV-2 Mpro and its complex with an α-ketoamide inhibitor. This was derived from a previously designed inhibitor but with the P3-P2 amide bond incorporated into a pyridone ring to enhance the half-life of the compound in plasma. On the basis of the unliganded structure, we developed the lead compound into a potent inhibitor of the SARS-CoV-2 Mpro The pharmacokinetic characterization of the optimized inhibitor reveals a pronounced lung tropism and suitability for administration by the inhalative route.


Asunto(s)
Amidas/química , Amidas/farmacología , Betacoronavirus/efectos de los fármacos , Betacoronavirus/enzimología , Cisteína Endopeptidasas/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/química , Amidas/metabolismo , Animales , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacocinética , Antivirales/farmacología , Sitios de Unión , Línea Celular Tumoral , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Cisteína Endopeptidasas/metabolismo , Diseño de Fármacos , Semivida , Humanos , Pulmón/metabolismo , Ratones , Modelos Moleculares , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacocinética , Dominios Proteicos , Multimerización de Proteína , Piridonas/química , SARS-CoV-2 , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA