Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Eur J Med Chem ; 256: 115463, 2023 Aug 05.
Artículo en Inglés | MEDLINE | ID: covidwho-2316659

RESUMEN

SARS-CoV-2 Omicron viruses possess a high antigenic shift, and the approved anti-SARS-CoV-2 drugs are extremely limited, which makes the development of new antiviral drugs for the clinical treatment and prevention of SARS-CoV-2 outbreaks imperative. We have previously discovered a new series of markedly potent small-molecule inhibitors of SARS-CoV-2 virus entry, exampled by the hit compound 2. Here, we report a further study of bioisosteric replacement of the eater linker at the C-17 position of 2 with a variety of aromatic amine moieties, followed by a focused structure-activity relationship study, leading to the discovery of a series of novel 3-O-ß-chacotriosyl BA amide derivatives as small-molecule Omicron fusion inhibitors with improved potency and selectivity index. Particularly, our medicinal chemistry efforts have resulted in a potent, and efficacious lead compound S-10 with appreciable pharmacokinetic properties, which exhibited broad-spectrum potency against Omicron and other variants with EC50 values ranging from 0.82 to 5.45 µM. Mutagenesis studies confirmed that inhibition of Omicron viral entry was mediated by the direct interaction with S in the prefusion state. These results reveal that S-10 is suitable for further optimization as Omicron fusion inhibitors, with the potential to be developed as therapeutic agents for the treatment and control of SARS-CoV-2 ant its variants infections.


Asunto(s)
Ácido Betulínico , COVID-19 , Humanos , SARS-CoV-2 , Amidas/farmacología , Aminas , Antirretrovirales
2.
Comb Chem High Throughput Screen ; 25(14): 2413-2428, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-2251463

RESUMEN

Favipiravir is a potential antiviral drug undergoing clinical trials to manage various viral infections, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Favipiravir possesses antiviral properties against RNA viruses, including SARS-CoV-2. Unfortunately, these viruses do not have authorized antiviral drugs for the management of diseases resulting from their infection, hence the dire need to accentuate the discovery of antiviral drugs that are efficacious and have a broad spectrum. Favipiravir acts primarily by blocking inward and outward movements of the virus from cells. Favipiravir is a prodrug undergoing intracellular phosphorylation and ribosylation to form an active form, favipiravir-RTP, which binds viral RNA-dependent RNA polymerase (RdRp). Considering the novel mechanism of favipiravir action, especially in managing viral infections, it is vital to pay more attention to the promised favipiravir hold in the management of SARS-CoV-2, its efficacy, and dosage regimen, and interactions with other drugs. In conclusion, favipiravir possesses antiviral properties against RNA viruses, including COVID- 19. Favipiravir is effective against SARS-CoV-2 infection through inhibition of RdRp. Pre-clinical and large-scalp prospective studies are recommended for efficacy and long-term safety of favipiravir in COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Virus , Humanos , SARS-CoV-2 , Estudios Prospectivos , Amidas/farmacología , Amidas/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , ARN Polimerasa Dependiente del ARN
3.
Molecules ; 28(1)2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: covidwho-2238921

RESUMEN

Although the incidence and mortality of SARS-CoV-2 infection has been declining during the pandemic, the problem related to designing novel antiviral drugs that could effectively resist viruses in the future remains relevant. As part of our continued search for chemical compounds that are capable of exerting an antiviral effect against the SARS-CoV-2 virus, we studied the ability of triterpenic acid amides to inhibit the SARS-CoV-2 main protease. Molecular modeling suggested that the compounds are able to bind to the active site of the main protease via non-covalent interactions. The FRET-based enzyme assay was used to reveal that compounds 1e and 1b can inhibit the SARS-CoV-2 main protease at micromolar concentrations.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Amidas/farmacología , Amidas/metabolismo , Antivirales/química , Inhibidores de Proteasas/química , Simulación del Acoplamiento Molecular
4.
Pharmacol Res Perspect ; 9(3): e00800, 2021 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1898944

RESUMEN

Antiprotozoal drug nitazoxanide (NTZ) has shown diverse pharmacological properties and has appeared in several clinical trials. Herein we present the synthesis, characterization, in vitro biological investigation, and in silico study of four hetero aryl amide analogs of NTZ. Among the synthesized molecules, compound 2 and compound 4 exhibited promising antibacterial activity against Escherichia coli (E. coli), superior to that displayed by the parent drug nitazoxanide as revealed from the in vitro antibacterial assay. Compound 2 displayed zone of inhibition of 20 mm, twice as large as the parent drug NTZ (10 mm) in their least concentration (12.5 µg/ml). Compound 1 also showed antibacterial effect similar to that of nitazoxanide. The analogs were also tested for in vitro cytotoxic activity by employing cell counting kit-8 (CCK-8) assay technique in HeLa cell line, and compound 2 was identified as a potential anticancer agent having IC50 value of 172 µg which proves it to be more potent than nitazoxanide (IC50  = 428 µg). Furthermore, the compounds were subjected to molecular docking study against various bacterial and cancer signaling proteins. The in vitro test results corroborated with the in silico docking study as compound 2 and compound 4 had comparatively stronger binding affinity against the proteins and showed a higher docking score than nitazoxanide toward human mitogen-activated protein kinase (MAPK9) and fatty acid biosynthesis enzyme (FabH) of E. coli. Moreover, the docking study demonstrated dihydrofolate reductase (DHFR) and thymidylate synthase (TS) as probable new targets for nitazoxanide and its synthetic analogs. Overall, the study suggests that nitazoxanide and its analogs can be a potential lead compound in the drug development.


Asunto(s)
Amidas , Antibacterianos , Antineoplásicos , Antiparasitarios , Nitrocompuestos , Tiazoles , Amidas/química , Amidas/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Antiparasitarios/química , Antiparasitarios/farmacología , Proteínas Bacterianas/metabolismo , Bioensayo , Supervivencia Celular/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Células HeLa , Humanos , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Nitrocompuestos/química , Nitrocompuestos/farmacología , Tetrahidrofolato Deshidrogenasa/metabolismo , Tiazoles/química , Tiazoles/farmacología , Timidilato Sintasa/metabolismo
5.
Mini Rev Med Chem ; 22(17): 2224-2236, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-1700911

RESUMEN

Among antiviral drugs, the vast majority targets only one or two related viruses. The conventional model, one virus - one drug, significantly limits therapeutic options. Therefore, in the strategy of controlling viral infections, there is a necessity to develop compounds with pleiotropic effects. Favipiravir (FPV) emerged as a strong candidate to become such a drug. The aim of the study is to present up-to-date information on the role of favipiravir in the treatment of viral respiratory infections. The anti-influenza activity of favipiravir has been confirmed in cell culture experiments, animal models, and clinical trials. Thoroughly different - from the previously registered drugs - mechanism of action suggests that FVP can be used as a countermeasure for the novel or re-emerging influenza virus infections. In recent months, favipiravir has been broadly investigated due to its potential efficacy in the treatment of COVID-19. Based on preclinical and clinical studies and a recently published meta-analysis it seems that favipiravir may be a promising antiviral drug in the treatment of patients with COVID-19. FPV is also effective against other RNA respiratory viruses and may be a candidate for the treatment of serious infections caused by human rhinovirus, respiratory syncytial virus, metapneumovirus, parainfluenza viruses and hantavirus pulmonary syndrome.


Asunto(s)
COVID-19 , Virus ARN , Virosis , Virus , Amidas/farmacología , Amidas/uso terapéutico , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Humanos , Pirazinas/farmacología , Pirazinas/uso terapéutico , Virosis/tratamiento farmacológico
6.
Molecules ; 27(3)2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: covidwho-1648677

RESUMEN

The human population is still facing appalling conditions due to several outbreaks of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) virus. The absence of specific drugs, appropriate vaccines for mutants, and knowledge of potential therapeutic agents makes this situation more difficult. Several 1, 2, 4-triazolo [1, 5-a] pyrimidine (TP)-derivative compounds were comprehensively studied for antiviral activities against RNA polymerase of HIV, HCV, and influenza viruses, and showed immense pharmacological interest. Therefore, TP-derivative compounds can be repurposed against the RNA-dependent RNA polymerase (RdRp) protein of SARS-CoV-2. In this study, a meta-analysis was performed to ensure the genomic variability and stability of the SARS-CoV-2 RdRp protein. The molecular docking of natural and synthetic TP compounds to RdRp and molecular dynamic (MD) simulations were performed to analyse the dynamic behaviour of TP compounds at the active site of the RdRp protein. TP compounds were also docked against other non-structural proteins (NSP1, NSP2, NSP3, NSP5, NSP8, NSP13, and NSP15) of SARS-CoV-2. Furthermore, the inhibition potential of TP compounds was compared with Remdesivir and Favipiravir drugs as a positive control. Additionally, TP compounds were analysed for inhibitory activity against SARS-CoV RdRp protein. This study demonstrates that TP analogues (monomethylated triazolopyrimidine and essramycin) represent potential lead molecules for designing an effective inhibitor to control viral replication. Furthermore, in vitro and in vivo studies will strengthen the use of these inhibitors as suitable drug candidates against SARS-CoV-2.


Asunto(s)
ARN Polimerasa Dependiente de ARN de Coronavirus/efectos de los fármacos , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , Pirimidinas/farmacología , Triazoles/farmacología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Amidas/farmacología , COVID-19/metabolismo , Dominio Catalítico/efectos de los fármacos , Biología Computacional/métodos , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Pirazinas/farmacología , Pirimidinas/química , ARN Viral/efectos de los fármacos , ARN Polimerasa Dependiente del ARN/efectos de los fármacos , ARN Polimerasa Dependiente del ARN/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , Triazoles/química , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
7.
Viruses ; 13(12)2021 12 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1572668

RESUMEN

Broad-spectrum antiviral therapies hold promise as a first-line defense against emerging viruses by blunting illness severity and spread until vaccines and virus-specific antivirals are developed. The nucleobase favipiravir, often discussed as a broad-spectrum inhibitor, was not effective in recent clinical trials involving patients infected with Ebola virus or SARS-CoV-2. A drawback of favipiravir use is its rapid clearance before conversion to its active nucleoside-5'-triphosphate form. In this work, we report a synergistic reduction of flavivirus (dengue, Zika), orthomyxovirus (influenza A), and coronavirus (HCoV-OC43 and SARS-CoV-2) replication when the nucleobases favipiravir or T-1105 were combined with the antimetabolite 6-methylmercaptopurine riboside (6MMPr). The 6MMPr/T-1105 combination increased the C-U and G-A mutation frequency compared to treatment with T-1105 or 6MMPr alone. A further analysis revealed that the 6MMPr/T-1105 co-treatment reduced cellular purine nucleotide triphosphate synthesis and increased conversion of the antiviral nucleobase to its nucleoside-5'-monophosphate, -diphosphate, and -triphosphate forms. The 6MMPr co-treatment specifically increased production of the active antiviral form of the nucleobases (but not corresponding nucleosides) while also reducing levels of competing cellular NTPs to produce the synergistic effect. This in-depth work establishes a foundation for development of small molecules as possible co-treatments with nucleobases like favipiravir in response to emerging RNA virus infections.


Asunto(s)
Antimetabolitos/farmacología , Antivirales/farmacología , Virus ARN/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Amidas/farmacología , Animales , Línea Celular , Sinergismo Farmacológico , Guanosina Trifosfato/metabolismo , Humanos , Metiltioinosina/farmacología , Mutación/efectos de los fármacos , Fosforribosil Pirofosfato/metabolismo , Pirazinas/farmacología , Virus ARN/clasificación , Virus ARN/genética , ARN Viral/efectos de los fármacos , ARN Viral/genética , Replicación Viral/efectos de los fármacos
8.
Sci Rep ; 11(1): 23670, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1560986

RESUMEN

Among cases of SARS-CoV-2 infections that result in serious conditions or death, many have pre-existing conditions such as hypertension and are on renin-angiotensin-aldosterone system (RAAS) inhibitors. The angiotensin-converting-enzyme-2 (ACE2), a key protein of the RAAS pathway, also mediates cellular entry of SARS-CoV-2. RAAS inhibitors might affect the expression levels of ace2, which could impact patient susceptibility to SARS-CoV-2. However, multi-organ-specific information is currently lacking and no species other than rodents have been examined. To address this knowledge gap, we treated adult zebrafish with the RAAS inhibitors aliskiren, olmesartan, and captopril for 7 consecutive days and performed qRT-PCR analysis of major RAAS pathway genes in the brain, gill, heart, intestine, kidney, and liver. Both olmesartan and captopril significantly increased ace2 expression in the heart, gill, and kidney. Olmesartan also increased ace2 expression in the intestine. Conversely, aliskiren significantly decreased ace2 expression in the heart. Discontinuation of compound treatments for 7 days did not return ace2 expression to baseline levels. While potential risks or benefits of antihypertensive RAAS inhibitors to SARS-CoV-2 infections in humans remain uncertain, this study provides new insights regarding the impact of RAAS inhibitors on organ-specific ace2 expression in another vertebrate model, thereby providing comparative data and laying scientific groundwork for future clinical decisions of RAAS inhibitor use in the context of COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Pez Cebra/metabolismo , Amidas/farmacología , Enzima Convertidora de Angiotensina 2/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , COVID-19/patología , COVID-19/virología , Fumaratos/farmacología , Branquias/efectos de los fármacos , Branquias/metabolismo , Humanos , Imidazoles/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Modelos Animales , SARS-CoV-2/aislamiento & purificación , Tetrazoles/farmacología
9.
Sci Rep ; 11(1): 23465, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1556248

RESUMEN

Human coronavirus NL63 (HCoV-NL63) mainly affects young children and immunocompromised patients, causing morbidity and mortality in a subset of patients. Since no specific treatment is available, this study aims to explore the anti-SARS-CoV-2 agents including favipiravir and remdesivir for treating HCoV-NL63 infection. We first successfully modelled the 3D structure of HCoV-NL63 RNA-dependent RNA polymerase (RdRp) based on the experimentally solved SARS-CoV-2 RdRp structure. Molecular docking indicated that favipiravir has similar binding affinities to SARS-CoV-2 and HCoV-NL63 RdRp with LibDock scores of 75 and 74, respectively. The LibDock scores of remdesivir to SARS-CoV-2 and HCoV-NL63 were 135 and 151, suggesting that remdesivir may have a higher affinity to HCoV-NL63 compared to SARS-CoV-2 RdRp. In cell culture models infected with HCoV-NL63, both favipiravir and remdesivir significantly inhibited viral replication and production of infectious viruses. Overall, remdesivir compared to favipiravir is more potent in inhibiting HCoV-NL63 in cell culture. Importantly, there is no evidence of resistance development upon long-term exposure to remdesivir. Furthermore, combining favipiravir or remdesivir with the clinically used antiviral cytokine interferon-alpha resulted in synergistic effects. These findings provided a proof-of-concept that anti-SARS-CoV-2 drugs, in particular remdesivir, have the potential to be repurposed for treating HCoV-NL63 infection.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Amidas/química , Antivirales/química , Coronavirus Humano NL63/enzimología , Pirazinas/química , ARN Polimerasa Dependiente del ARN/química , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología , Alanina/química , Alanina/metabolismo , Alanina/farmacología , Amidas/metabolismo , Amidas/farmacología , Animales , Antivirales/metabolismo , Antivirales/farmacología , Sitios de Unión , Técnicas de Cultivo de Célula , Línea Celular , Coronavirus Humano NL63/fisiología , Haplorrinos , Humanos , Simulación del Acoplamiento Molecular , Pirazinas/metabolismo , Pirazinas/farmacología , ARN Polimerasa Dependiente del ARN/metabolismo , Replicación Viral/efectos de los fármacos
10.
Mikrochim Acta ; 188(12): 434, 2021 11 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1536308

RESUMEN

A novel and sensitive voltammetric nanosensor was developed for the first time for trace level monitoring of favipiravir based on gold/silver core-shell nanoparticles (Au@Ag CSNPs) with conductive polymer poly (3,4-ethylene dioxythiophene) polystyrene sulfonate (PEDOT:PSS) and functionalized multi carbon nanotubes (F-MWCNTs) on a glassy carbon electrode (GCE). The formation of Au@Ag CSNPs/PEDOT:PSS/F-MWCNT composite was confirmed by various analytical techniques, including X-ray diffraction (XRD), ultraviolet-visible spectroscopy (UV-Vis), transmission electron microscopy (TEM), energy-dispersive X-ray spectroscopy (EDX), and field-emission scanning electron microscopy (SEM). Under the optimized conditions and at a typical working potential of + 1.23 V (vs. Ag/AgCl), the Au@Ag CSNPs/PEDOT:PSS/F-MWCNT/GCE revealed linear quantitative ranges from 0.005 to 0.009 and 0.009 to 1.95 µM with a limit of detection 0.46 nM (S/N = 3) with acceptable relative standard deviations (1.1-4.9 %) for pharmaceutical formulations, urine, and human plasma samples without applying any sample pretreatment (1.12-4.93%). The interference effect of antiviral drugs, biological compounds, and amino acids was negligible, and the sensing system demonstrated outstanding reproducibility, repeatability, stability, and reusability. The findings revealed that this assay strategy has promising applications in diagnosing FAV in clinical samples, which could be attributed to the large surface area on active sites and high conductivity of bimetallic nanocomposite.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Electroquímica/métodos , Nanopartículas del Metal/química , Nanocompuestos/química , Nanomedicina/métodos , Nanotecnología/métodos , Pirazinas/farmacología , Coloides/química , Electrodos , Oro/química , Humanos , Límite de Detección , Modelos Lineales , Nanotubos , Polímeros/química
11.
ChemMedChem ; 16(22): 3418-3427, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: covidwho-1525425

RESUMEN

Currently, limited therapeutic options are available for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). We have developed a set of pyrazine-based small molecules. A series of pyrazine conjugates was synthesized by microwave-assisted click chemistry and benzotriazole chemistry. All the synthesized conjugates were screened against the SAR-CoV-2 virus and their cytotoxicity was determined. Computational studies were carried out to validate the biological data. Some of the pyrazine-triazole conjugates (5 d-g) and (S)-N-(1-(benzo[d]thiazol-2-yl)-2-phenylethyl)pyrazine-2-carboxamide 12 i show significant potency against SARS-CoV-2 among the synthesized conjugates. The selectivity index (SI) of potent conjugates indicates significant efficacy compared to the reference drug (Favipiravir).


Asunto(s)
Antivirales/farmacología , Pirazinas/farmacología , SARS-CoV-2/efectos de los fármacos , Amidas/farmacología , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Antivirales/toxicidad , Chlorocebus aethiops , ARN Polimerasa Dependiente de ARN de Coronavirus/metabolismo , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Molecular , Pirazinas/síntesis química , Pirazinas/metabolismo , Pirazinas/toxicidad , Relación Estructura-Actividad Cuantitativa , Células Vero
12.
Int J Mol Sci ; 22(21)2021 Oct 21.
Artículo en Inglés | MEDLINE | ID: covidwho-1480798

RESUMEN

Disseminated intravascular coagulation (DIC) is a severe condition characterized by the systemic formation of microthrombi complicated with bleeding tendency and organ dysfunction. In the last years, it represents one of the most frequent consequences of coronavirus disease 2019 (COVID-19). The pathogenesis of DIC is complex, with cross-talk between the coagulant and inflammatory pathways. The objective of this study is to investigate the anti-inflammatory action of ultramicronized palmitoylethanolamide (um-PEA) in a lipopolysaccharide (LPS)-induced DIC model in rats. Experimental DIC was induced by continual infusion of LPS (30 mg/kg) for 4 h through the tail vein. Um-PEA (30 mg/kg) was given orally 30 min before and 1 h after the start of intravenous infusion of LPS. Results showed that um-PEA reduced alteration of coagulation markers, as well as proinflammatory cytokine release in plasma and lung samples, induced by LPS infusion. Furthermore, um-PEA also has the effect of preventing the formation of fibrin deposition and lung damage. Moreover, um-PEA was able to reduce the number of mast cells (MCs) and the release of its serine proteases, which are also necessary for SARS-CoV-2 infection. These results suggest that um-PEA could be considered as a potential therapeutic approach in the management of DIC and in clinical implications associated to coagulopathy and lung dysfunction, such as COVID-19.


Asunto(s)
Amidas/uso terapéutico , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Coagulación Intravascular Diseminada/tratamiento farmacológico , Etanolaminas/uso terapéutico , Ácidos Palmíticos/uso terapéutico , Sepsis/complicaciones , Amidas/química , Amidas/farmacología , Animales , Trastornos de la Coagulación Sanguínea/etiología , COVID-19/patología , COVID-19/virología , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Coagulación Intravascular Diseminada/etiología , Etanolaminas/química , Etanolaminas/farmacología , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Lipopolisacáridos/toxicidad , Pulmón/metabolismo , Pulmón/patología , Masculino , Mastocitos/citología , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ácidos Palmíticos/química , Ácidos Palmíticos/farmacología , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , Ratas , Ratas Sprague-Dawley , SARS-CoV-2/aislamiento & purificación , Sepsis/patología , Serina Proteasas/metabolismo
13.
Sci Rep ; 11(1): 19998, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1462031

RESUMEN

Understanding the effects of metabolism on the rational design of novel and more effective drugs is still a considerable challenge. To the best of our knowledge, there are no entirely computational strategies that make it possible to predict these effects. From this perspective, the development of such methodologies could contribute to significantly reduce the side effects of medicines, leading to the emergence of more effective and safer drugs. Thereby, in this study, our strategy is based on simulating the electron ionization mass spectrometry (EI-MS) fragmentation of the drug molecules and combined with molecular docking and ADMET models in two different situations. In the first model, the drug is docked without considering the possible metabolic effects. In the second model, each of the intermediates from the EI-MS results is docked, and metabolism occurs before the drug accesses the biological target. As a proof of concept, in this work, we investigate the main antiviral drugs used in clinical research to treat COVID-19. As a result, our strategy made it possible to assess the biological activity and toxicity of all potential by-products. We believed that our findings provide new chemical insights that can benefit the rational development of novel drugs in the future.


Asunto(s)
Antivirales/metabolismo , Tratamiento Farmacológico de COVID-19 , Descubrimiento de Drogas , SARS-CoV-2/efectos de los fármacos , Adenina/efectos adversos , Adenina/análogos & derivados , Adenina/metabolismo , Adenina/farmacología , Adenosina/efectos adversos , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacología , Adenosina Monofosfato/efectos adversos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/farmacología , Alanina/efectos adversos , Alanina/análogos & derivados , Alanina/metabolismo , Alanina/farmacología , Amidas/efectos adversos , Amidas/metabolismo , Amidas/farmacología , Antivirales/efectos adversos , Antivirales/farmacología , COVID-19/metabolismo , Cloroquina/efectos adversos , Cloroquina/análogos & derivados , Cloroquina/metabolismo , Cloroquina/farmacología , Diseño de Fármacos , Humanos , Redes y Vías Metabólicas , Simulación del Acoplamiento Molecular , Nitrocompuestos/efectos adversos , Nitrocompuestos/metabolismo , Nitrocompuestos/farmacología , Pirazinas/efectos adversos , Pirazinas/metabolismo , Pirazinas/farmacología , Pirrolidinas/efectos adversos , Pirrolidinas/metabolismo , Pirrolidinas/farmacología , Ribavirina/efectos adversos , Ribavirina/metabolismo , Ribavirina/farmacología , SARS-CoV-2/metabolismo , Tiazoles/efectos adversos , Tiazoles/metabolismo , Tiazoles/farmacología
14.
J Am Coll Cardiol ; 78(16): 1635-1654, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: covidwho-1454219

RESUMEN

Coronavirus disease-2019 (COVID-19) is associated with systemic inflammation, endothelial activation, and multiorgan manifestations. Lipid-modulating agents may be useful in treating patients with COVID-19. These agents may inhibit viral entry by lipid raft disruption or ameliorate the inflammatory response and endothelial activation. In addition, dyslipidemia with lower high-density lipoprotein cholesterol and higher triglyceride levels portend worse outcomes in patients with COVID-19. Upon a systematic search, 40 randomized controlled trials (RCTs) with lipid-modulating agents were identified, including 17 statin trials, 14 omega-3 fatty acids RCTs, 3 fibrate RCTs, 5 niacin RCTs, and 1 dalcetrapib RCT for the management or prevention of COVID-19. From these 40 RCTs, only 2 have reported preliminary results, and most others are ongoing. This paper summarizes the ongoing or completed RCTs of lipid-modulating agents in COVID-19 and the implications of these trials for patient management.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/prevención & control , Ácidos Grasos Omega-3/uso terapéutico , Ácidos Fíbricos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Niacina/uso terapéutico , Amidas/farmacología , Amidas/uso terapéutico , Ésteres/farmacología , Ésteres/uso terapéutico , Ácidos Grasos Omega-3/farmacología , Ácidos Fíbricos/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Reguladores del Metabolismo de Lípidos/farmacología , Reguladores del Metabolismo de Lípidos/uso terapéutico , Niacina/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Compuestos de Sulfhidrilo/farmacología , Compuestos de Sulfhidrilo/uso terapéutico
15.
EBioMedicine ; 72: 103595, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: covidwho-1433162

RESUMEN

BACKGROUND: Favipiravir and Molnupiravir, orally available antivirals, have been reported to exert antiviral activity against SARS-CoV-2. First efficacy data have been recently reported in COVID-19 patients. METHODS: We here report on the combined antiviral effect of both drugs in a SARS-CoV-2 Syrian hamster infection model. The infected hamsters were treated twice daily with the vehicle (the control group) or a suboptimal dose of each compound or a combination of both compounds. FINDINGS: When animals were treated with a combination of suboptimal doses of Molnupiravir and Favipiravir at the time of infection, a marked combined potency at endpoint is observed. Infectious virus titers in the lungs of animals treated with the combination are reduced by ∼5 log10 and infectious virus are no longer detected in the lungs of >60% of treated animals. When start of treatment was delayed with one day a reduction of titers in the lungs of 2.4 log10 was achieved. Moreover, treatment of infected animals nearly completely prevented transmission to co-housed untreated sentinels. Both drugs result in an increased mutation frequency of the remaining viral RNA recovered from the lungs of treated animals. In the combo-treated hamsters, an increased frequency of C-to-T mutations in the viral RNA is observed as compared to the single treatment groups which may explain the pronounced antiviral potency of the combination. INTERPRETATION: Our findings may lay the basis for the design of clinical studies to test the efficacy of the combination of Molnupiravir/Favipiravir in the treatment of COVID-19. FUNDING: stated in the acknowledgment.


Asunto(s)
Amidas/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Citidina/análogos & derivados , Hidroxilaminas/uso terapéutico , Pulmón/virología , Pirazinas/uso terapéutico , Amidas/farmacología , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/transmisión , Citidina/farmacología , Citidina/uso terapéutico , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Hidroxilaminas/farmacología , Mesocricetus , Pirazinas/farmacología , ARN Viral , Resultado del Tratamiento , Carga Viral
16.
FASEB J ; 35(6): e21651, 2021 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1388031

RESUMEN

The SARS-CoV-2 pandemic imposed a large burden on health and society. Therapeutics targeting different components and processes of the viral infection replication cycle are being investigated, particularly to repurpose already approved drugs. Spike protein is an important target for both vaccines and therapeutics. Insights into the mechanisms of spike-ACE2 binding and cell fusion could support the identification of compounds with inhibitory effects. Here, we demonstrate that the integrity of disulfide bonds within the receptor-binding domain (RBD) plays an important role in the membrane fusion process although their disruption does not prevent binding of spike protein to ACE2. Several reducing agents and thiol-reactive compounds are able to inhibit viral entry. N-acetyl cysteine amide, L-ascorbic acid, JTT-705, and auranofin prevented syncytia formation, viral entry into cells, and infection in a mouse model, supporting disulfides of the RBD as a therapeutically relevant target.


Asunto(s)
Acetilcisteína/análogos & derivados , Amidas/farmacología , Ácido Ascórbico/farmacología , Auranofina/farmacología , Tratamiento Farmacológico de COVID-19 , COVID-19 , Disulfuros/metabolismo , Ésteres/farmacología , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Compuestos de Sulfhidrilo/farmacología , Internalización del Virus/efectos de los fármacos , Acetilcisteína/farmacología , COVID-19/metabolismo , COVID-19/patología , Células HEK293 , Humanos
17.
Nat Commun ; 12(1): 1735, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: covidwho-1387332

RESUMEN

Despite no or limited pre-clinical evidence, repurposed drugs are massively evaluated in clinical trials to palliate the lack of antiviral molecules against SARS-CoV-2. Here we use a Syrian hamster model to assess the antiviral efficacy of favipiravir, understand its mechanism of action and determine its pharmacokinetics. When treatment is initiated before or simultaneously to infection, favipiravir has a strong dose effect, leading to reduction of infectious titers in lungs and clinical alleviation of the disease. Antiviral effect of favipiravir correlates with incorporation of a large number of mutations into viral genomes and decrease of viral infectivity. Antiviral efficacy is achieved with plasma drug exposure comparable with those previously found during human clinical trials. Notably, the highest dose of favipiravir tested is associated with signs of toxicity in animals. Thereby, pharmacokinetic and tolerance studies are required to determine whether similar effects can be safely achieved in humans.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Pirazinas/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , COVID-19/virología , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Femenino , Genoma Viral , Pulmón/virología , Mesocricetus , SARS-CoV-2/genética , Células Vero , Carga Viral/efectos de los fármacos
18.
Biochemistry ; 59(48): 4601-4608, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1387100

RESUMEN

The development of reliable ways of predicting the binding free energies of covalent inhibitors is a challenge for computer-aided drug design. Such development is important, for example, in the fight against the SARS-CoV-2 virus, in which covalent inhibitors can provide a promising tool for blocking Mpro, the main protease of the virus. This work develops a reliable and practical protocol for evaluating the binding free energy of covalent inhibitors. Our protocol presents a major advance over other approaches that do not consider the chemical contribution of the binding free energy. Our strategy combines the empirical valence bond method for evaluating the reaction energy profile and the PDLD/S-LRA/ß method for evaluating the noncovalent part of the binding process. This protocol has been used in the calculations of the binding free energy of an α-ketoamide inhibitor of Mpro. Encouragingly, our approach reproduces the observed binding free energy. Our study of covalent inhibitors of cysteine proteases indicates that in the choice of an effective warhead it is crucial to focus on the exothermicity of the point on the free energy surface of a peptide cleavage that connects the acylation and deacylation steps. Overall, we believe that our approach should provide a powerful and effective method for in silico design of covalent drugs.


Asunto(s)
Amidas/química , Amidas/farmacología , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , SARS-CoV-2/enzimología , Amidas/metabolismo , Simulación del Acoplamiento Molecular , Péptido Hidrolasas/química , Inhibidores de Proteasas/metabolismo , Unión Proteica , Conformación Proteica , SARS-CoV-2/efectos de los fármacos , Termodinámica
19.
FEBS Lett ; 595(18): 2366-2382, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1363633

RESUMEN

Favipiravir is a broad-spectrum inhibitor of viral RNA-dependent RNA polymerase (RdRp) currently being used to manage COVID-19. Accumulation of mutations in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RdRp may facilitate antigenic drift, generating favipiravir resistance. Focussing on the chain-termination mechanism utilized by favipiravir, we used high-throughput interface-based protein design to generate > 100 000 designs of the favipiravir-binding site of RdRp and identify mutational hotspots. We identified several single-point mutants and designs having a sequence identity of 97%-98% with wild-type RdRp, suggesting that SARS-CoV-2 can develop favipiravir resistance with few mutations. Out of 134 mutations documented in the CoV-GLUE database, 63 specific mutations were already predicted as resistant in our calculations, thus attaining ˜ 47% correlation with the sequencing data. These findings improve our understanding of the potential signatures of adaptation in SARS-CoV-2 against favipiravir.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Pirazinas/farmacología , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/genética , Farmacorresistencia Viral/genética , Mutación/genética , Mutación Puntual/genética
20.
Biochem Biophys Res Commun ; 571: 26-31, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: covidwho-1312941

RESUMEN

The pandemic of SARS-CoV-2 has necessitated expedited research efforts towards finding potential antiviral targets and drug development measures. While new drug discovery is time consuming, drug repurposing has been a promising area for elaborate virtual screening and identification of existing FDA approved drugs that could possibly be used for targeting against functions of various proteins of SARS-CoV-2 virus. RNA dependent RNA polymerase (RdRp) is an important enzyme for the virus that mediates replication of the viral RNA. Inhibition of RdRp could inhibit viral RNA replication and thus new virus particle production. Here, we screened non-nucleoside antivirals and found three out of them to be strongest in binding to RdRp out of which two retained binding even using molecular dynamic simulations. We propose these two drugs as potential RdRp inhibitors which need further in-depth testing.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , ARN Polimerasa Dependiente de ARN de Coronavirus/antagonistas & inhibidores , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , Amidas/farmacología , Antivirales/química , Bencimidazoles/farmacología , COVID-19/virología , Carbamatos/farmacología , Dominio Catalítico , Simulación por Computador , ARN Polimerasa Dependiente de ARN de Coronavirus/química , Ciclopropanos/farmacología , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Fluorenos/farmacología , Humanos , Lactamas Macrocíclicas/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Pandemias , Prolina/análogos & derivados , Prolina/farmacología , Conformación Proteica , Quinoxalinas/farmacología , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA