Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 536
Filtrar
1.
SAR QSAR Environ Res ; 32(11): 863-888, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34634208

RESUMO

The novel severe acute respiratory syndrome coronavirus (SARS CoV-2) was introduced as an epidemic in 2019 and had millions of deaths worldwide. Given the importance of this disease, the recommendation and design of new active compounds are crucial. 3-chymotrypsin-like protease (3 CLpro) inhibitors have been identified as potent compounds for treating SARS-CoV-2 disease. So, the design of new 3 CLpro inhibitors was proposed using a quantitative structure-activity relationship (QSAR) study. In this context, a powerful adaptive least absolute shrinkage and selection operator (ALASSO) penalized variable selection method with inherent advantages coupled with a nonlinear artificial neural network (ANN) modelling method were used to provide a QSAR model with high interpretability and predictability. After evaluating the accuracy and validity of the developed ALASSO-ANN model, new compounds were proposed using effective descriptors, and the biological activity of the new compounds was predicted. Ligand-receptor (LR) interactions were also performed to confirm the interaction strength of the compounds using molecular docking (MD) study. The pharmacokinetics properties and calculated Lipinski's rule of five were applied to all proposed compounds. Due to the ease of synthesis of these suggested new compounds, it is expected that they have acceptable pharmacological properties.


Assuntos
Antivirais/química , Proteases 3C de Coronavírus/antagonistas & inibidores , Inibidores de Proteases/química , SARS-CoV-2/efeitos dos fármacos , Antivirais/farmacocinética , Proteases 3C de Coronavírus/química , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Redes Neurais de Computação , Inibidores de Proteases/farmacocinética , Relação Quantitativa Estrutura-Atividade , Reprodutibilidade dos Testes , SARS-CoV-2/enzimologia
2.
J Med Chem ; 64(16): 11972-11989, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34405680

RESUMO

The three pan-genotypic HCV NS3/4A protease inhibitors (PIs) currently in clinical use-grazoprevir, glecaprevir, and voxilaprevir-are quinoxaline-based P2-P4 macrocycles and thus exhibit similar resistance profiles. Using our quinoxaline-based P1-P3 macrocyclic lead compounds as an alternative chemical scaffold, we explored structure-activity relationships (SARs) at the P2 and P4 positions to develop pan-genotypic PIs that avoid drug resistance. A structure-guided strategy was used to design and synthesize two series of compounds with different P2 quinoxalines in combination with diverse P4 groups of varying sizes and shapes, with and without fluorine substitutions. Our SAR data and cocrystal structures revealed the interplay between the P2 and P4 groups, which influenced inhibitor binding and the overall resistance profile. Optimizing inhibitor interactions in the S4 pocket led to PIs with excellent antiviral activity against clinically relevant PI-resistant HCV variants and genotype 3, providing potential pan-genotypic inhibitors with improved resistance profiles.


Assuntos
Antivirais/uso terapêutico , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Compostos Macrocíclicos/uso terapêutico , Inibidores de Proteases/uso terapêutico , Quinoxalinas/uso terapêutico , Animais , Antivirais/síntese química , Antivirais/metabolismo , Antivirais/farmacocinética , Cristalografia por Raios X , Farmacorresistência Viral/efeitos dos fármacos , Compostos Macrocíclicos/síntese química , Compostos Macrocíclicos/metabolismo , Compostos Macrocíclicos/farmacocinética , Masculino , Estrutura Molecular , Inibidores de Proteases/síntese química , Inibidores de Proteases/metabolismo , Inibidores de Proteases/farmacocinética , Ligação Proteica , Quinoxalinas/síntese química , Quinoxalinas/metabolismo , Quinoxalinas/farmacocinética , Ratos Sprague-Dawley , Serina Proteases/metabolismo , Relação Estrutura-Atividade , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo
3.
Future Med Chem ; 13(17): 1435-1450, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34169728

RESUMO

The COVID-19 outbreak has thrown the world into an unprecedented crisis. It has posed a challenge to scientists around the globe who are working tirelessly to combat this pandemic. We herein report a set of molecules that may serve as possible inhibitors of the SARS-CoV-2 main protease. To identify these molecules, we followed a combinatorial structure-based strategy, which includes high-throughput virtual screening, molecular docking and WaterMap calculations. The study was carried out using Protein Data Bank structures 5R82 and 6Y2G. DrugBank, Enamine, Natural product and Specs databases, along with a few known antiviral drugs, were used for the screening. WaterMap analysis aided in the recognition of high-potential molecules that can efficiently displace binding-site waters. This study may help the discovery and development of antiviral drugs against SARS-CoV-2.


Assuntos
Antivirais/química , Antivirais/farmacologia , Proteases 3C de Coronavírus/química , Inibidores de Proteases/química , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/enzimologia , Antivirais/farmacocinética , Antivirais/uso terapêutico , Sítios de Ligação/efeitos dos fármacos , Catálise , Simulação por Computador , Bases de Dados Factuais , Ensaios de Triagem em Larga Escala , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Inibidores de Proteases/farmacocinética , Termodinâmica , Água/química
4.
J Med Chem ; 64(12): 8076-8100, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34081466

RESUMO

The beta-site APP cleaving enzyme 1, known as BACE1, has been a widely pursued Alzheimer's disease drug target owing to its critical role in the production of amyloid-beta. We have previously reported the clinical development of LY2811376 and LY2886721. LY2811376 advanced to Phase I before development was terminated due to nonclinical retinal toxicity. LY2886721 advanced to Phase II, but development was halted due to abnormally elevated liver enzymes. Herein, we report the discovery and clinical development of LY3202626, a highly potent, CNS-penetrant, and low-dose BACE inhibitor, which successfully addressed these key development challenges.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Compostos Heterocíclicos com 2 Anéis/farmacologia , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Pirróis/farmacologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Barreira Hematoencefálica/fisiologia , Encéfalo/metabolismo , Cristalografia por Raios X , Cães , Estabilidade de Medicamentos , Compostos Heterocíclicos com 2 Anéis/síntese química , Compostos Heterocíclicos com 2 Anéis/farmacocinética , Humanos , Células Madin Darby de Rim Canino , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Inibidores de Proteases/síntese química , Inibidores de Proteases/metabolismo , Inibidores de Proteases/farmacocinética , Ligação Proteica , Pirazinas/síntese química , Pirazinas/farmacocinética , Pirróis/síntese química , Pirróis/farmacocinética , Ratos , Relação Estrutura-Atividade
5.
J Med Chem ; 64(4): 1873-1888, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33588527

RESUMO

Accumulation of amyloid ß peptides (Aß) is thought to be one of the causal factors of Alzheimer's disease (AD). The aspartyl protease ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) is the rate-limiting protease for Aß production, and therefore, BACE1 inhibition is a promising therapeutic approach for the treatment of AD. Starting with a dihydro-1,3-thiazine-based lead, Compound J, we discovered atabecestat 1 (JNJ-54861911) as a centrally efficacious BACE1 inhibitor that was advanced into the EARLY Phase 2b/3 clinical trial for the treatment of preclinical AD patients. Compound 1 demonstrated robust and dose-dependent Aß reduction and showed sufficient safety margins in preclinical models. The potential of reactive metabolite formation was evaluated in a covalent binding study to assess its irreversible binding to human hepatocytes. Unfortunately, the EARLY trial was discontinued due to significant elevation of liver enzymes, and subsequent analysis of the clinical outcomes showed dose-related cognitive worsening.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Inibidores de Proteases/uso terapêutico , Piridinas/uso terapêutico , Tiazinas/uso terapêutico , Peptídeos beta-Amiloides/metabolismo , Animais , Cães , Canal de Potássio ERG1/antagonistas & inibidores , Término Precoce de Ensaios Clínicos , Feminino , Humanos , Masculino , Camundongos , Inibidores de Proteases/síntese química , Inibidores de Proteases/farmacocinética , Piridinas/síntese química , Piridinas/farmacocinética , Ratos Sprague-Dawley , Tiazinas/síntese química , Tiazinas/farmacocinética
6.
Bioconjug Chem ; 32(2): 279-289, 2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33523652

RESUMO

Reducing the required frequence of drug dosing can improve the adherence of patients to chronic treatments. Hence, drugs with longer in vivo half-lives are highly desirable. One of the most promising approaches to extend the in vivo half-life of drugs is conjugation to human serum albumin (HSA). In this work, we describe the use of AlbuBinder 1, a small-molecule noncovalent HSA binder, to extend the in vivo half-life and pharmacology of small-molecule BMP1/TLL inhibitors in humanized mice (HSA KI/KI). A series of conjugates of AlbuBinder 1 with BMP1/TLL inhibitors were prepared. In particular, conjugate c showed good solubility and a half-life extension of >20-fold versus the parent molecule in the HSA KI/KI mice, reaching half-lives of >48 h with maintained maximal inhibition of plasma BMP1/TLL. The same conjugate showed a half-life of only 3 h in the wild-type mice, suggesting that the half-life extension was principally due to specific interactions with HSA. It is envisioned that conjugation to AlbuBinder 1 should be applicable to a wide range of small molecule or peptide drugs with short half-lives. In this context, AlbuBinders represent a viable alternative to existing half-life extension technologies.


Assuntos
Metaloproteases/metabolismo , Inibidores de Proteases/farmacologia , Albumina Sérica Humana/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Animais , Proteína Morfogenética Óssea 1/metabolismo , Meia-Vida , Humanos , Camundongos , Estudo de Prova de Conceito , Inibidores de Proteases/farmacocinética
7.
Mol Divers ; 25(3): 1827-1838, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33400085

RESUMO

Main protease (Mpro) of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) intervenes in the replication and transcription processes of the virus. Hence, it is a lucrative target for anti-viral drug development. In this study, molecular modeling analyses were performed on the structure activity data of recently reported diverse SARS-CoV-2 Mpro inhibitors to understand the structural requirements for higher inhibitory activity. The classification-based quantitative structure-activity relationship (QSAR) models were generated between SARS-CoV-2 Mpro inhibitory activities and different descriptors. Identification of structural fingerprints to increase or decrease in the inhibitory activity was mapped for possible inclusion/exclusion of these fingerprints in the lead optimization process. Challenges in ADME properties of protease inhibitors were also discussed to overcome the problems of oral bioavailability. Further, depending on the modeling results, we have proposed novel as well as potent SARS-CoV-2 Mpro inhibitors.


Assuntos
Proteases 3C de Coronavírus/antagonistas & inibidores , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , SARS-CoV-2/enzimologia , Disponibilidade Biológica , Proteases 3C de Coronavírus/química , Modelos Moleculares , Inibidores de Proteases/farmacocinética , Conformação Proteica , SARS-CoV-2/efeitos dos fármacos , Relação Estrutura-Atividade
8.
Viruses ; 13(1)2021 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-33451132

RESUMO

In 2019 an outbreak occurred which resulted in a global pandemic. The causative agent has been identified in a virus belonging to theCoronaviridae family, similar to the agent of SARS, referred to as SARS-CoV-2. This epidemic spread rapidly globally with high morbidity and mortality. Although vaccine development is at a very advanced stage, there are currently no truly effective antiviral drugs to treat SARS-CoV-2 infection. In this study we present systematic and integrative antiviral drug repurposing effort aimed at identifying, among the drugs already authorized for clinical use, some active inhibitors of the SARS-CoV-2 main protease. The most important result of this analysis is the demonstration that ethacrynic acid, a powerful diuretic, is revealed to be an effective inhibitor of SARS-CoV-2 main protease. Even with all the necessary cautions, given the particular nature of this drug, these data can be the starting point for the development of an effective therapeutic strategy against SARS-CoV-2.


Assuntos
Antivirais/farmacologia , Proteases 3C de Coronavírus/antagonistas & inibidores , Ácido Etacrínico/farmacologia , Inibidores de Proteases/farmacocinética , SARS-CoV-2/efeitos dos fármacos , Antivirais/química , Domínio Catalítico , Proteases 3C de Coronavírus/química , Proteases 3C de Coronavírus/metabolismo , Bases de Dados Factuais , Reposicionamento de Medicamentos , Ácido Etacrínico/química , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Inibidores de Proteases/química , SARS-CoV-2/enzimologia
9.
Eur J Pharmacol ; 890: 173648, 2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33069672

RESUMO

In an attempt to search for selective inhibitors against the SARS-CoV-2 which caused devastating of lives and livelihoods across the globe, 415 natural metabolites isolated from several plants, fungi and bacteria, belonging to different classes, were investigated. The drug metabolism and safety profiles were computed in silico and the results showed seven compounds namely fusaric acid, jasmonic acid, jasmonic acid methyl ester, putaminoxin, putaminoxin B and D, and stagonolide K were predicted to having considerable absorption, metabolism, distribution and excretion parameters (ADME) and safety indices. Molecular docking against the receptor binding domain (RBD) of spike glycoprotein (S1) and the main protease (Mpro) exposed the compounds having better binding affinity to main protease as compared to the S1 receptor binding domain. The docking results were compared to an antiviral drug penciclovir reportedly of clinical significance in treating the SARS-CoV-2 infected patients. The results demonstrated the test compounds jasmonic acid, putaminoxins B and D bound to the HIS-CYS catalytic dyad as well as to other residues within the MPro active site with much greater affinity than penciclovir. The findings of the study suggest that these compounds could be explored as potential SARS-CoV-2 inhibitors, and could further be combined with the experimental investigations to develop effective therapeutics to deal with the present pandemic.


Assuntos
Antivirais/farmacologia , Produtos Biológicos/farmacologia , Proteases 3C de Coronavírus/metabolismo , Compostos Fitoquímicos/farmacologia , Inibidores de Proteases/farmacologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Antivirais/farmacocinética , Bactérias/metabolismo , Produtos Biológicos/farmacocinética , Barreira Hematoencefálica/metabolismo , Proteases 3C de Coronavírus/antagonistas & inibidores , Ciclopentanos/farmacocinética , Ciclopentanos/farmacologia , Fungos/metabolismo , Humanos , Absorção Intestinal , Lactonas/farmacocinética , Lactonas/farmacologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Oxilipinas/farmacocinética , Oxilipinas/farmacologia , Compostos Fitoquímicos/farmacocinética , Plantas/metabolismo , Inibidores de Proteases/farmacocinética , Ligação Proteica , Domínios Proteicos , SARS-CoV-2
10.
Spectrochim Acta A Mol Biomol Spectrosc ; 244: 118825, 2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-32866803

RESUMO

Novel antiviral active molecule 2- [(4,6-diaminopyrimidin-2-yl)sulfanyl]-N-(4-fluoro- phenyl)acetamide has been synthesised and characterized by FT-IR and FT-Raman spectra. The equilibrium geometry, natural bond orbital calculations and vibrational assignments have been carried out using density functional B3LYP method with the 6-311G++(d,p) basis set. The complete vibrational assignments for all the vibrational modes have been supported by normal coordinate analysis, force constants and potential energy distributions. A detailed analysis of the intermolecular interactions has been performed based on the Hirshfeld surfaces. Drug likeness has been carried out based on Lipinski's rule and the absorption, distribution, metabolism, excretion and toxicity of the title molecule has been calculated. Antiviral potency of 2- [(4,6-diaminopyrimidin-2-yl)sulfanyl]-N-(4-fluoro-phenyl) acetamide has been investigated by docking against SARS-CoV-2 protein. The optimized geometry shows near-planarity between the phenyl ring and the pyrimidine ring. Differences in the geometries due to the substitution of the most electronegative fluorine atom and intermolecular contacts due to amino pyrimidine were analyzed. NBO analysis reveals the formation of two strong stable hydrogen bonded N-H···N intermolecular interactions and weak intramolecular interactions C-H···O and N-H···O. The Hirshfeld surfaces and consequently the 2D-fingerprint confirm the nature of intermolecular interactions and their quantitative contributions towards the crystal packing. The red shift in N-H stretching frequency exposed from IR substantiate the formation of N-H···N intermolecular hydrogen bond. Drug likeness and absorption, distribution, metabolism, excretion and toxicity properties analysis gives an idea about the pharmacokinetic properties of the title molecule. The binding energy -8.7 kcal/mol of the nonbonding interaction present a clear view that 2- [(4,6-diaminopyrimidin-2-yl)sulfanyl]-N-(4-fluoro- phenyl) acetamide can irreversibly interact with SARS-CoV-2 protease.


Assuntos
Acetamidas/química , Antivirais/química , Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Pandemias , Pneumonia Viral/tratamento farmacológico , Inibidores de Proteases/química , Pirimidinas/química , Proteínas não Estruturais Virais/antagonistas & inibidores , Acetamidas/farmacocinética , Antivirais/farmacocinética , Betacoronavirus/enzimologia , COVID-19 , Proteases 3C de Coronavírus , Cristalografia por Raios X , Cisteína Endopeptidases , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Estrutura Molecular , Dinâmica não Linear , Inibidores de Proteases/farmacocinética , Conformação Proteica , Pirimidinas/farmacocinética , Teoria Quântica , SARS-CoV-2 , Espectroscopia de Infravermelho com Transformada de Fourier , Análise Espectral Raman , Termodinâmica , Vibração , Tratamento Farmacológico da COVID-19
11.
J Proteome Res ; 20(1): 1015-1026, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33350309

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) expresses a multifunctional papain-like proteinase (PLpro), which mediates the processing of the viral replicase polyprotein. Inhibition of PLpro has been shown to suppress the viral replication. This study aimed to explore new anti-PLpro candidates by applying virtual screening based on GRL0617, a known PLpro inhibitor of SARS coronavirus (SARS-CoV). The three-dimensional (3D) structure of SARS-CoV-2 PLpro was built by homology modeling, using SARS-CoV PLpro as the template. The model was refined and studied through molecular dynamic simulation. AutoDock Vina was then used to perform virtual screening where 50 chemicals with at least 65% similarity to GRL0617 were docked with the optimized SARS-CoV-2 PLpro. In this screening, 5-(aminomethyl)-2-methyl-N-[(1R)-1-naphthalen-1-ylethyl]benzamide outperformed GRL0617 in terms of binding affinity (-9.7 kcal/mol). Furthermore, 2-(4-fluorobenzyl)-5-nitro-1H-isoindole-1,3(2H)-dione (previously introduced as an inhibitor of cyclooxygenase-2), 3-nitro-N-[(1r)-1-phenylethyl]-5-(trifluoromethyl)benzamide (inhibitor against Mycobacterium tuberculosis), as well as the recently introduced SARS-CoV-2 PLpro inhibitor 5-acetamido-2-methyl-N-[(1S)-1-naphthalen-1-ylethyl]benzamide showed promising affinity for the viral proteinase. All of the identified compounds demonstrated an acceptable pharmacokinetic profile. In conclusion, our findings represent rediscovery of analgesic, anti-inflammatory, antibacterial, or antiviral drugs as promising pharmaceutical candidates against the ongoing coronavirus.


Assuntos
Antivirais/farmacologia , Proteases 3C de Coronavírus/antagonistas & inibidores , Proteases 3C de Coronavírus/química , Inibidores de Proteases/farmacologia , Antivirais/efeitos adversos , Antivirais/química , Antivirais/farmacocinética , Sítios de Ligação , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Simulação por Computador , Proteases 3C de Coronavírus/genética , Proteases 3C de Coronavírus/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Microssomos Hepáticos/efeitos dos fármacos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Inibidores de Proteases/efeitos adversos , Inibidores de Proteases/química , Inibidores de Proteases/farmacocinética , Conformação Proteica , Relação Estrutura-Atividade
12.
J Mol Graph Model ; 101: 107758, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33007575

RESUMO

A novel strain of coronavirus, namely, SARS-CoV-2 identified in Wuhan city of China in December 2019, continues to spread at a rapid rate worldwide. There are no specific therapies available and investigations regarding the treatment of this disease are still lacking. In order to identify a novel potent inhibitor, we performed blind docking studies on the main virus protease Mpro with eight approved drugs belonging to four pharmacological classes such as: anti-malarial, anti-bacterial, anti-infective and anti-histamine. Among the eight studied compounds, Lymecycline and Mizolastine appear as potential inhibitors of this protease. When docked against Mpro crystal structure, these two compounds revealed a minimum binding energy of -8.87 and -8.71 kcal/mol with 168 and 256 binding modes detected in the binding substrate pocket, respectively. Further, to study the interaction mechanism and conformational dynamics of protein-ligand complexes, Molecular dynamic simulation and MM/PBSA binding free calculations were performed. Our results showed that both Lymecycline and Mizolastine bind in the active site. And exhibited good binding affinities towards target protein. Moreover, the ADMET analysis also indicated drug-likeness properties. Thus it is suggested that the identified compounds can inhibit Chymotrypsin-like protease (3CLpro) of SARS-CoV-2.


Assuntos
Cisteína Endopeptidases/química , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Animais , Antibacterianos/química , Antivirais/química , Antivirais/farmacocinética , Antivirais/farmacologia , Sítios de Ligação , Simulação por Computador , Proteases 3C de Coronavírus , Cisteína Endopeptidases/metabolismo , Bases de Dados de Produtos Farmacêuticos , Aprovação de Drogas , Reposicionamento de Medicamentos , Antagonistas dos Receptores Histamínicos/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Inibidores de Proteases/farmacocinética , Proteínas não Estruturais Virais/metabolismo
13.
Bioorg Chem ; 103: 104201, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32890999

RESUMO

Alzheimer's disease (AD) is a neurodegenerative process that compromises cognitive functions. The physiopathology of AD is multifactorial and is mainly supported by the cholinergic and amyloid hypotheses, which allows the identification the fundamental role of some markers, such as the enzymes acetylcholinesterase (AChE) and ß-secretase (BACE-1), and the ß-amyloid peptide (Aß). In this work, we prepared a series of chalcones and 2'-aminochalcones, which were tested against AChE and BACE-1 enzymes and on the aggregation of Aß. All compounds inhibited AChE activity with different potencies. We have found that the majority of chalcones having the amino group are able to inhibit BACE-1, which was not observed for chalcones without this group. The most active compound is the one derived from 2,3-dichlorobenzaldeyde, having an IC50 value of 2.71 µM. A molecular docking study supported this result, showing a good interaction of the amino group with aspartic acid residues of the catalytic diade of BACE-1. Thioflavin-T fluorescence emission is reduced in 30 - 40%, when Aß42 is incubated in the presence of some chalcones under aggregation conditions. In vitro cytotoxicity and in silico prediction of pharmacokinetic properties were also conducted in this study.


Assuntos
Chalconas/farmacologia , Inibidores da Colinesterase/farmacologia , Inibidores de Proteases/farmacologia , Acetilcolinesterase/metabolismo , Doença de Alzheimer/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Linhagem Celular Tumoral , Chalconas/síntese química , Chalconas/metabolismo , Chalconas/farmacocinética , Chlorocebus aethiops , Inibidores da Colinesterase/síntese química , Inibidores da Colinesterase/metabolismo , Inibidores da Colinesterase/farmacocinética , Electrophorus , Humanos , Camundongos , Simulação de Acoplamento Molecular , Fragmentos de Peptídeos/metabolismo , Inibidores de Proteases/síntese química , Inibidores de Proteases/farmacocinética , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Células Vero
14.
Curr Top Med Chem ; 20(24): 2146-2167, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32621718

RESUMO

BACKGROUND: The vast geographical expansion of novel coronavirus and an increasing number of COVID-19 affected cases have overwhelmed health and public health services. Artificial Intelligence (AI) and Machine Learning (ML) algorithms have extended their major role in tracking disease patterns, and in identifying possible treatments. OBJECTIVE: This study aims to identify potential COVID-19 protease inhibitors through shape-based Machine Learning assisted by Molecular Docking and Molecular Dynamics simulations. METHODS: 31 Repurposed compounds have been selected targeting the main coronavirus protease (6LU7) and a machine learning approach was employed to generate shape-based molecules starting from the 3D shape to the pharmacophoric features of their seed compound. Ligand-Receptor Docking was performed with Optimized Potential for Liquid Simulations (OPLS) algorithms to identify highaffinity compounds from the list of selected candidates for 6LU7, which were subjected to Molecular Dynamic Simulations followed by ADMET studies and other analyses. RESULTS: Shape-based Machine learning reported remdesivir, valrubicin, aprepitant, and fulvestrant as the best therapeutic agents with the highest affinity for the target protein. Among the best shape-based compounds, a novel compound identified was not indexed in any chemical databases (PubChem, Zinc, or ChEMBL). Hence, the novel compound was named 'nCorv-EMBS'. Further, toxicity analysis showed nCorv-EMBS to be suitable for further consideration as the main protease inhibitor in COVID-19. CONCLUSION: Effective ACE-II, GAK, AAK1, and protease 3C blockers can serve as a novel therapeutic approach to block the binding and attachment of the main COVID-19 protease (PDB ID: 6LU7) to the host cell and thus inhibit the infection at AT2 receptors in the lung. The novel compound nCorv- EMBS herein proposed stands as a promising inhibitor to be evaluated further for COVID-19 treatment.


Assuntos
Betacoronavirus/efeitos dos fármacos , Betacoronavirus/enzimologia , Infecções por Coronavirus/tratamento farmacológico , Pneumonia Viral/tratamento farmacológico , Inibidores de Proteases/farmacologia , Algoritmos , COVID-19 , Mineração de Dados , Bases de Dados Factuais , Reposicionamento de Medicamentos , Humanos , Ligantes , Aprendizado de Máquina , Modelos Teóricos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Pandemias , Inibidores de Proteases/química , Inibidores de Proteases/farmacocinética , SARS-CoV-2
15.
Biomolecules ; 10(6)2020 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-32599963

RESUMO

The pandemic associated with Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV2) and its disease named COVID-19 challenged the scientific community to discover effective therapeutic solutions in a short period. Repurposing existing drugs is one viable approach that emphasizes speed during these urgent times. Famotidine, a class A G protein-coupled receptor antagonist used for the treatment of gastroesophageal reflux was recently identified in an in silico screening. Additionally, a recent retrospective clinical report showed that the treatment with famotidine provided a good outcome in patients infected with SARS-CoV2. A clinical trial testing effectiveness of famotidine in combination with hydroxychloroquine is currently ongoing in the United States (US). In the 1990s, famotidine was described as an antiviral agent against human immunodeficiency virus (HIV). Interestingly, some HIV protease inhibitors are presently being used against SARS-CoV2. However, it is not clear if famotidine could be effective against SARS-CoV2. Thus, by using a computational analysis, we aimed to examine if the antiviral effect of famotidine could be related to the inhibition of proteases involved in the virus replication. Our results showed that famotidine could interact within the catalytic site of the three proteases associated with SARS-CoV2 replication. However, weak binding affinity of famotidine to these proteases suggests that a successful famotidine therapy could likely be achieved only in combination with other antiviral drugs. Finally, analysis of famotidine's pharmacokinetic parameters indicated that its effect against SARS-CoV2 infection could be reached only upon intravenous administration. This work will contribute to the pharmacological knowledge of famotidine as an antiviral agent against SARS-CoV2.


Assuntos
Antivirais/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Famotidina/uso terapêutico , Pneumonia Viral/tratamento farmacológico , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Administração Intravenosa , Antivirais/administração & dosagem , Antivirais/farmacocinética , COVID-19 , Simulação por Computador , Reposicionamento de Medicamentos , Famotidina/administração & dosagem , Famotidina/farmacocinética , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Pandemias , Inibidores de Proteases/administração & dosagem , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/uso terapêutico , Replicação Viral/efeitos dos fármacos
16.
Bioorg Chem ; 101: 103992, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32554279

RESUMO

Thiazole derivatives are known to possess various biological activities such as antiparasitic, antifungal, antimicrobial and antiproliferative activities. Matrix metalloproteinases (MMPs) are important protease target involved in tumor progression including angiogenesis, tissue invasion, and migration. Therefore, MMPs have also been reported as potential diagnostic and prognostic biomarkers in many types of cancer. Herein, new aryl thiazoles were synthesized and evaluated for their anticancer effects on a panel of cancer cell lines including the invasive MDA-MB-231 line. Some of these compounds showed IC50 values in the submicromolar range in anti-proliferative assays. In order to examine the relationship between their anticancer activity and MMPs targets, the compounds were evaluated for their inhibitory effects on MMP-2 and 9. That data obtained revealed that most of these compounds were potent dual MMP-2/9 inhibitors at nanomolar concentrations. Among these, 2-(1-(2-(2-((E)-4-iodobenzylidene)hydrazineyl)-4-methylthiazol-5-yl)ethylidene)hydrazine-1-carboximidamide (4a) was the most potent non-selective dual MMP-2/9 inhibitor with inhibitory concentrations of 56 and 38 nM respectively. When compound 4a was tested in an MDA-MB-231, HCT-116, MCF-7 model, it effectively inhibited tumor growth, strongly induced cancer cell apoptosis, inhibit cell migration, and suppressed cell cycle progression leading to DNA fragmentation. Taken together, the results of our studies indicate that the newly discovered thiazole-based MMP-2/9 inhibitors have significant potential for anticancer treatment.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Descoberta de Drogas , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Triazóis/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Inibidores de Proteases/química , Inibidores de Proteases/farmacocinética , Relação Estrutura-Atividade , Cicatrização/efeitos dos fármacos
17.
Eur J Pharmacol ; 880: 173171, 2020 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-32437743

RESUMO

Cathepsin S (CatS) is a cysteine protease found in lysosomes of hematopoietic and microglial cells and in secreted form in the extracellular space. While CatS has been shown to contribute significantly to neuropathic pain, the precise mechanisms remain unclear. In this report, we describe JNJ-39641160, a novel non-covalent, potent, selective and orally-available CatS inhibitor that is peripherally restricted (non-CNS penetrant) and may represent an innovative class of immunosuppressive and analgesic compounds and tools useful toward investigating peripheral mechanisms of CatS in neuropathic pain. In C57BL/6 mice, JNJ-39641160 dose-dependently blocked the proteolysis of the invariant chain, and inhibited both T-cell activation and antibody production to a vaccine antigen. In the spared nerve injury (SNI) model of chronic neuropathic pain, in which T-cell activation has previously been demonstrated to be a prerequisite for the development of pain hypersensitivity, JNJ-39641160 fully reversed tactile allodynia in wild-type mice but was completely ineffective in the same model in CatS knockout mice (which exhibited a delayed onset in allodynia). By contrast, in the acute mild thermal injury (MTI) model, JNJ-39641160 only weakly attenuated allodynia at the highest dose tested. These findings support the hypothesis that blockade of peripheral CatS alone is sufficient to fully reverse allodynia following peripheral nerve injury and suggest that the mechanism of action likely involves interruption of T-cell activation and peripheral cytokine release. In addition, they provide important insights toward the development of selective CatS inhibitors for the treatment of neuropathic pain in humans.


Assuntos
Analgésicos/uso terapêutico , Catepsinas/antagonistas & inibidores , Hiperalgesia/tratamento farmacológico , Imunossupressores/uso terapêutico , Neuralgia/tratamento farmacológico , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Analgésicos/farmacocinética , Analgésicos/farmacologia , Animais , Encéfalo/metabolismo , Catepsinas/genética , Catepsinas/metabolismo , Linhagem Celular , Citocinas/imunologia , Temperatura Alta , Humanos , Hiperalgesia/imunologia , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Imunossupressores/farmacocinética , Imunossupressores/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/imunologia , Traumatismos dos Nervos Periféricos/imunologia , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/farmacologia , Nervo Isquiático/lesões , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Toxoide Tetânico/administração & dosagem , Tato
18.
Science ; 368(6489): 409-412, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32198291

RESUMO

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.


Assuntos
Amidas/química , Amidas/farmacologia , Betacoronavirus/efeitos dos fármacos , Betacoronavirus/enzimologia , Cisteína Endopeptidases/química , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Proteínas não Estruturais Virais/química , Amidas/metabolismo , Animais , Antivirais/química , Antivirais/metabolismo , Antivirais/farmacocinética , Antivirais/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , Proteases 3C de Coronavírus , Cristalografia por Raios X , Cisteína Endopeptidases/metabolismo , Desenho de Fármacos , Meia-Vida , Humanos , Pulmão/metabolismo , Camundongos , Modelos Moleculares , Inibidores de Proteases/metabolismo , Inibidores de Proteases/farmacocinética , Domínios Proteicos , Multimerização Proteica , Piridonas/química , SARS-CoV-2 , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos
19.
Mol Pharm ; 17(3): 933-943, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32011889

RESUMO

The prostate-specific membrane antigen (PSMA) is a type II transmembrane glycoprotein that is highly expressed in the malignant human prostate epithelium. Therefore, PSMA has emerged as a very attractive target for developing radiopharmaceuticals for the diagnosis, e.g., by positron emission tomography (PET) imaging, and radiotherapy of prostate cancer. The aim of this study was to develop 18F-labeled PSMA ligands bearing different 18F-glycosyl moieties to study the effect on the in vivo clearance behavior of radiotracers in addition to their tumor binding ability. Therefore, we applied click chemistry-based 18F-fluoroglcosylation using 2-deoxy-2-[18F]fluoroglucosyl azide or 6-deoxy-6-[18F]fluoroglucosyl azide as prosthetic groups for the radiosynthesis of the 18F-fluoroglycosylated glutamate-urea-lysine-based PSMA inhibitors 2-[18F]FGlc-PSMA ([18F]7) and 6-[18F]FGlc-PSMA ([18F]8). The PSMA inhibitory potencies were determined by competitive radioligand binding assays using 99mTc-MIP-1404 and PSMA-expressing PC-3 PIP cells, revealing moderate PSMA inhibitory potencies for [18F]7 (IC50 = 234 nM) and [18F]8 (IC50 = 59 nM). Biodistribution and small-animal PET studies were performed using PSMA-positive PC-3 PIP and PSMA-negative PC-3 tumor-bearing nude mice. PSMA inhibitors [18F]7 and [18F]8 were obtained in high radioactivity yields of 19-22% (nondecay-corrected, referred to [18F]fluoride) and with molar activities of 71-136 GBq/µmol. In the biodistribution studies, the uptake levels of [18F]7 and [18F]8 in PC-3 PIP tumors were 13 ± 3%ID/g and 6 ± 5%ID/g at 60 min p.i., respectively. PSMA-negative PC-3 tumors and all other tissues had negligible low uptake values. Interestingly, [18F]7 had high uptake in the kidneys, with remarkable retention from 30 to 60 min p.i. (74 to 72%ID/g). In contrast, [18F]8 revealed a low uptake of 7.5%ID/g in the kidneys at 30 min p.i. and was rapidly cleared through the kidney (0.9%ID/g at 120 min p.i.). In direct comparison to a 68Ga-PSMA-11 PET scan of the same mouse, [18F]7 and [18F]8 showed 2- to 3-fold higher uptake values in PC-3 PIP tumors. Both radiotracers were solely cleared via the kidneys and not via the hepatobiliary pathway. The regional kidney distribution pattern of the tracers in the kidneys revealed that 68Ga-PSMA-11 and 2-[18F]FGlc-PSMA([18F]7) mainly accumulated in the cortex of the kidneys, whereas 6-[18F]FGlc-PSMA([18F]8) showed a 10-fold lower kidney uptake with accumulation in the inner medulla or pelvis of the kidneys. Overall, the developed 6-fluoroglucosyl derivative [18F]8, with its considerably low kidney uptake and fast clearance, demonstrated high uptake in PSMA-positive tumors in vivo. This candidate could, therefore, be valuable for translation into the clinic.


Assuntos
Antígenos de Superfície/metabolismo , Radioisótopos de Flúor/química , Glutamato Carboxipeptidase II/metabolismo , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/metabolismo , Inibidores de Proteases/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Animais , Química Click/métodos , Modelos Animais de Doenças , Feminino , Glutamato Carboxipeptidase II/antagonistas & inibidores , Xenoenxertos , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Ligantes , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Nus , Células PC-3 , Tomografia por Emissão de Pósitrons , Inibidores de Proteases/química , Compostos Radiofarmacêuticos/química , Distribuição Tecidual
20.
Ther Drug Monit ; 42(2): 229-244, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32004247

RESUMO

To date, therapeutic drug monitoring (TDM) has played an important role in the management of pregnant HIV patients on highly active antiretroviral therapy. Historically, in pregnant women living with HIV, the third agent in triple therapy has been either non-nucleoside reverse transcriptase inhibitors or protease inhibitors (PIs). PIs have been the preferred agents because of their robustness from the perspective of viral resistance and the dominant drug class for the management of HIV during pregnancy for the previous decade. As with many drugs used during pregnancy, pharmacokinetic changes decrease exposure to these agents as the pregnancy progresses. This can lead to viral escape at the time of pregnancy and ultimately increase the risk of mother-to-child transmission (MTCT) of HIV. TDM has been well-established for this class of highly active antiretroviral therapy, and appropriate dose adjustment studies have been performed. At present, there is a shift from the traditional treatment paradigm in pregnancy to a new drug class, integrase strand transfer inhibitors (INSTIs). Although INSTIs are affected by pharmacokinetic changes during pregnancy, they do not harbor the same issues with viral escape as seen with PIs at birth and in general eliminate the need for boosting with additional agents like ritonavir (r) and cobicistat (c) [bar elvitegravir (EVG)] that can lead to interactions with treatment of other common infections in HIV, including tuberculosis. Furthermore, INSTIs are the most successful medication for rapidly reducing the viral load (VL) in HIV patients, a useful factor where VL may be unknown, or in late presenters. These merits make INSTIs the best choice in pregnancy, although their use has been hindered in recent years by a report of neural tube defects from a large African study with dolutegravir (DTG). New data from Botswana and Brazil indicate that this risk is less significant than previously reported, necessitating further data to shed light on this critical issue. Current international guidelines including DHHS, EACS, WHO, and BHIVA (for patients with VLs >100,000 copies/mL or late presenters) now recommend INSTIs as first-line agents. The role of TDM in INSTIs shifts to cases of insufficient viral suppression with standard adherence measures, cases of drug-drug interactions, or cases where EVG/c is continued throughout pregnancy, and thus remains an important aspect of HIV care in pregnancy.


Assuntos
Antirretrovirais/uso terapêutico , Terapia Antirretroviral de Alta Atividade/métodos , Monitoramento de Medicamentos/métodos , Infecções por HIV/tratamento farmacológico , Inibidores de Integrase de HIV/uso terapêutico , Complicações Infecciosas na Gravidez/tratamento farmacológico , Antirretrovirais/farmacocinética , Área Sob a Curva , Aleitamento Materno , Interações Medicamentosas , Feminino , Inibidores de Integrase de HIV/farmacocinética , Humanos , Taxa de Depuração Metabólica , Gravidez , Inibidores de Proteases/farmacocinética , Inibidores de Proteases/uso terapêutico , Inibidores da Transcriptase Reversa/farmacocinética , Inibidores da Transcriptase Reversa/uso terapêutico , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...