Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Publication year range
1.
Québec; INESSS; 17 mars 2022.
Non-conventional in French | BRISA/RedTESA | ID: biblio-1367473

ABSTRACT

CONTEXTE: Le présent document ainsi que les constats qu'il énonce ont été rédigés en réponse à une interpellation du ministère de la Santé et des Services sociaux dans le contexte de la crise sanitaire liée à la maladie à coronavirus (COVID-19) au Québec. L'objectif est de réaliser une recension des données publiées et de mobiliser les savoirs clés afin d'informer les décideurs publics et les professionnels de la santé et des services sociaux. Bien que les constats reposent sur un repérage exhaustif des données scientifiques publiées, sur l'évaluation de la qualité méthodologique des études et sur une appréciation du niveau de preuve scientifique par paramètre clinique d'intérêt ainsi que sur la consultation de cliniciens avec différentes spécialités et expertises, le processus ne repose pas entièrement sur les normes habituelles à l'INESSS. Dans les circonstances d'une telle crise de santé publique, l'INESSS reste à l'affût de toutes nouvelles données, qu'elles soient de nature scientifique ou contextuelle, susceptibles de lui faire modifier cette réponse. PRÉSENTATION DE LA DEMANDE: La COVID-19 est une maladie causée par le virus SRAS-CoV-2 qui infecte préférentiellement les cellules du tractus respiratoire. Le SRAS-CoV-2 est un virus à ARN qui se lie aux cellules humaines par le biais de ses glycoprotéines S à l'enzyme de conversion de l'angiotensine 2 (de l'anglais ACE 2) [Letko et al., 2020]. Cette enzyme se trouve notamment à la surface des cellules épithéliales qui tapissent le tractus respiratoire, le tube digestif, les reins et le cœur [Zou et al., 2020; Li et al., 2003]. Compte tenu de la transmissibilité accrue du SRAS-CoV-2 comparativement au SRAS-CoV, des chercheurs ont émis l'hypothèse que d'autres glycoprotéines de surface contenant de l'acide sialique pourraient permettre aux particules vi


Subject(s)
Humans , Ritonavir/therapeutic use , 3C Viral Proteases/antagonists & inhibitors , SARS-CoV-2/drug effects , COVID-19/drug therapy , Health Evaluation , Cost-Benefit Analysis
2.
Microbiol Spectr ; 9(3): e0102521, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34787443

ABSTRACT

The fatal pathogen enterovirus 71 (EV71) is a major cause of hand-foot-and-mouth disease (HFMD), which leads to serious neurological syndromes. While there are no effective clinical agents available for EV71 treatment thus far, EV71 3C protease (3Cpro), a cysteine protease encoded by the virus, has become a promising drug target for discovery of antiviral drugs, given that it plays a crucial role in virus proliferation and interferes with host cell function. Here, we report two inhibitors of EV71 3Cpro, FOPMC and FIOMC, that were developed from previously reported cyanohydrin derivative (R)-1 by replacing the acyl cyanohydrin group with 4-iminooxazolidin-2-one. FOPMC and FIOMC have potent antiviral activity and dramatically improved metabolic stability. These two inhibitors demonstrated broad anti-EV effects on various cell lines and five epidemic viral strains. We further illuminated the binding models between 3Cpro and FOPMC/FIOMC through molecular docking and molecular dynamics simulations. The substitution of an acyl cyanohydrin group with 4-iminooxazolidin-2-one does make FOPMC and FIOMC potent anti-EV71 drug candidates as universal nonclassical bioisosteres with a cyanohydrin moiety. IMPORTANCE EV71 is one of the most epidemic agents of HFMD. Thus far, there are no antiviral drugs available for clinical usage. The conserved EV71 3Cpro plays pivotal roles in virus proliferation and defense host immunity, as well as having no homology in host cells, making it a most promising antiviral target. In this work, we identified that propyl- and isopropyl-substituted 4-iminooxazolidin-2-one moieties (FOPMC and FIOMC) effectively inhibited five epidemic viral strains in rhabdomyosarcoma (RD), HEK-293T, and VeroE6 cell lines. The inhibition mechanism was also illustrated with molecular docking and molecular dynamics (MD) simulations. The successful replacement of the labile cyanohydrin greatly improved the stability and pharmacokinetic properties of (R)-1, making 4-iminooxazolidin-2-one a nonclassical bioisosteric moiety of cyanohydrin. This discovery addressed a critical issue of the primitive structural scaffold of these promising anti-EV71 inhibitors and could lead to their development as broad-spectrum anti-EV agents.


Subject(s)
3C Viral Proteases , Antiviral Agents , Enterovirus A, Human , Virus Replication , Animals , Humans , 3C Viral Proteases/antagonists & inhibitors , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line, Tumor , Chlorocebus aethiops , Enterovirus A, Human/drug effects , Enterovirus A, Human/growth & development , Hand, Foot and Mouth Disease/drug therapy , Hand, Foot and Mouth Disease/prevention & control , Hand, Foot and Mouth Disease/virology , HEK293 Cells , Molecular Docking Simulation , Molecular Dynamics Simulation , Nitriles/chemistry , Nitriles/pharmacology , Vero Cells , Virus Replication/drug effects
3.
Viruses ; 13(11)2021 10 21.
Article in English | MEDLINE | ID: mdl-34834926

ABSTRACT

Foot-and-mouth-disease virus (FMDV) is a picornavirus that causes a highly contagious disease of cloven-hoofed animals resulting in economic losses worldwide. The 3C protease (3Cpro) is the main protease essential in the picornavirus life cycle, which is an attractive antiviral target. Here, we used computer-aided virtual screening to filter potential anti-FMDV agents from the natural phytochemical compound libraries. The top 23 filtered compounds were examined for anti-FMDV activities by a cell-based assay, two of which possessed antiviral effects. In the viral and post-viral entry experiments, luteolin and isoginkgetin could significantly block FMDV growth with low 50% effective concentrations (EC50). Moreover, these flavonoids could reduce the viral load as determined by RT-qPCR. However, their prophylactic activities were less effective. Both the cell-based and the fluorescence resonance energy transfer (FRET)-based protease assays confirmed that isoginkgetin was a potent FMDV 3Cpro inhibitor with a 50% inhibition concentration (IC50) of 39.03 ± 0.05 and 65.3 ± 1.7 µM, respectively, whereas luteolin was less effective. Analyses of the protein-ligand interactions revealed that both compounds fit in the substrate-binding pocket and reacted to the key enzymatic residues of the 3Cpro. Our findings suggested that luteolin and isoginkgetin are promising antiviral agents for FMDV and other picornaviruses.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , Antiviral Agents/pharmacology , Biflavonoids/pharmacology , Enzyme Inhibitors/pharmacology , Foot-and-Mouth Disease Virus/drug effects , Foot-and-Mouth Disease Virus/enzymology , Foot-and-Mouth Disease/virology , Luteolin/pharmacology , 3C Viral Proteases/chemistry , 3C Viral Proteases/genetics , 3C Viral Proteases/metabolism , Animals , Antiviral Agents/chemistry , Biflavonoids/chemistry , Computer Simulation , Enzyme Inhibitors/chemistry , Foot-and-Mouth Disease Virus/chemistry , Foot-and-Mouth Disease Virus/genetics , Humans , Luteolin/chemistry , Phytochemicals/chemistry , Phytochemicals/pharmacology
4.
Int J Mol Sci ; 22(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34830015

ABSTRACT

Coronaviruses cause diseases in humans and livestock. The SARS-CoV-2 is infecting millions of human beings, with high morbidity and mortality worldwide. The main protease (Mpro) of coronavirus plays a pivotal role in viral replication and transcription, which, in theory, is an attractive drug target for antiviral drug development. It has been extensively discussed whether Xanthohumol is able to help COVID-19 patients. Here, we report that Xanthohumol, a small molecule in clinical trials from hops (Humulus lupulus), was a potent pan-inhibitor for various coronaviruses by targeting Mpro, for example, betacoronavirus SARS-CoV-2 (IC50 value of 1.53 µM), and alphacoronavirus PEDV (IC50 value of 7.51 µM). Xanthohumol inhibited Mpro activities in the enzymatical assays, while pretreatment with Xanthohumol restricted the SARS-CoV-2 and PEDV replication in Vero-E6 cells. Therefore, Xanthohumol is a potent pan-inhibitor of coronaviruses and an excellent lead compound for further drug development.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , Flavonoids/chemistry , Propiophenones/chemistry , Protease Inhibitors/chemistry , SARS-CoV-2/enzymology , 3C Viral Proteases/chemistry , 3C Viral Proteases/metabolism , Alphacoronavirus/enzymology , Alphacoronavirus/physiology , Amino Acid Sequence , Animals , Binding Sites , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Biological Products/therapeutic use , COVID-19/virology , Catalytic Domain , Chlorocebus aethiops , Coronavirus/enzymology , Coronavirus/physiology , Flavonoids/metabolism , Flavonoids/pharmacology , Flavonoids/therapeutic use , Humans , Molecular Docking Simulation , Propiophenones/metabolism , Propiophenones/pharmacology , Propiophenones/therapeutic use , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , SARS-CoV-2/isolation & purification , Sequence Alignment , Vero Cells , Virus Replication/drug effects , COVID-19 Drug Treatment
5.
Sci Rep ; 11(1): 18970, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556768

ABSTRACT

It is widely accepted that Hepatitis A virus (HAV) is responsible for liver failure and even death in older people and in people with other serious health issues; so, proposing new compounds with inhibitory activity can help to treated of these disease's. In current study, a new class of quinolines is proposed with inhibitor activity of the HAV proteinase. So, in the first step, fused quinoline derivatives has been synthesized in short reaction time (12.0 min) and high efficiency yields (94%) in presence of 1-carboxymethyl-2,3-dimethylimidazolium iodide ([cmdmim]I) ionic liquid catalyst using a new method. In the following, chemical reactivity and inhibitory activity of synthesized quinolines were evaluated in density functional theory (DFT) framework and molecular docking methodologies. High global softness (0.67 eV), low HOMOSWBNNT-LUMO4a gap (4.78 eV), and more negative adsorption energy (- 87.9 kJ mol-1) in these quinolines reveal that the 4a and 4b compounds have better delivery than other quinolines using SWBNNT as suitable carrier to target cells. Molecular docking shows that the best cavity of the HAV has - 134.2 kJ mol-1 interaction energy involving bonding and non-bonding interactions. In fact, these interactions are between fused quinolines with especial geometries and sidechain flexibility amino acids residues inside the best binding site of the HAV, as hydrogen bonding, steric, and electrostatic interactions. So, these interactions imply that proposed fused quinolines have good inhibitor activity for the HAV.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , Antiviral Agents/pharmacology , Hepatitis A/drug therapy , Quinolines/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Hepatitis A/virology , Hepatitis A virus/drug effects , Hepatitis A virus/enzymology , Humans , Molecular Docking Simulation , Molecular Structure , Quinolines/chemistry , Quinolines/therapeutic use , Structure-Activity Relationship
6.
Bioorg Med Chem ; 42: 116219, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34077853

ABSTRACT

Covalent target modulation with small molecules has been emerging as a promising strategy for drug discovery. However, covalent inhibitory antibody remains unexplored due to the lack of efficient strategies to engineer antibody with desired bioactivity. Herein, we developed an intracellular selection method to generate covalent inhibitory antibody against human rhinovirus 14 (HRV14) 3C protease through unnatural amino acid mutagenesis along the heavy chain complementarity-determining region 3 (CDR-H3). A library of antibody mutants was thus constructed and screened in vivo through co-expression with the target protease. Using this screening strategy, six covalent antibodies with proximity-enabled bioactivity were identified, which were shown to covalently target HRV14-3C protease with high inhibitory potency and exquisite selectivity. Compared to structure-based rational design, this library-based screening method provides a simple and efficient way for the discovery and engineering of covalent antibody for enzyme inhibition.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , Antibodies/pharmacology , Complementarity Determining Regions/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Rhinovirus/enzymology , 3C Viral Proteases/metabolism , Antibodies/chemistry , Cysteine Proteinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
7.
Antiviral Res ; 192: 105102, 2021 08.
Article in English | MEDLINE | ID: mdl-34082057

ABSTRACT

As one of the principal etiological agents of hand, foot, and mouth disease (HFMD), enterovirus 71 (EV71) is associated with severe neurological complications or fatal diseases, while without effective medications thus far. Here we applied dually activated Michael acceptor to develop a series of reversible covalent compounds for EV71 3C protease (3Cpro), a promising antiviral drug target that plays an essential role during viral replication by cleaving the precursor polyprotein, inhibiting host protein synthesis, and evading innate immunity. Among them, cyanoacrylate and Boc-protected cyanoarylamide derivatives (SLQ-4 and SLQ-5) showed effective antiviral activity against EV71. The two inhibitors exhibited broad antiviral effects, acting on RD, 293T, and Vero cell lines, as well as on EV71 A, B, C, CVA16, and CVB3 viral strains. We further determined the binding pockets between the two inhibitors and 3Cpro based on docking studies. These results, together with our previous studies, provide evidence to elucidate the mechanism of action of these two reversible covalent inhibitors and contribute to the development of clinically effective medicines to treat EV71 infections.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Protease Inhibitors/pharmacology , 3C Viral Proteases/chemistry , Acrylamides/chemistry , Acrylamides/pharmacology , Animals , Antiviral Agents/chemistry , Cell Line , Cell Survival/drug effects , Cyanoacrylates/chemistry , Cyanoacrylates/pharmacology , Enterovirus/classification , Enterovirus/drug effects , Enterovirus Infections/virology , Humans , Molecular Docking Simulation , Protease Inhibitors/chemistry , Virus Replication/drug effects
8.
Angew Chem Int Ed Engl ; 60(24): 13294-13301, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33749121

ABSTRACT

The generation of bioactive molecules from inactive precursors is a crucial step in the chemical evolution of life, however, mechanistic insights into this aspect of abiogenesis are scarce. Here, we investigate the protein-catalyzed formation of antivirals by the 3C-protease of enterovirus D68. The enzyme induces aldol condensations yielding inhibitors with antiviral activity in cells. Kinetic and thermodynamic analyses reveal that the bioactivity emerges from a dynamic reaction system including inhibitor formation, alkylation of the protein target by the inhibitors, and competitive addition of non-protein nucleophiles to the inhibitors. The most active antivirals are slowly reversible inhibitors with elongated target residence times. The study reveals first examples for the chemical evolution of bio-actives through protein-catalyzed, non-enzymatic C-C couplings. The discovered mechanism works under physiological conditions and might constitute a native process of drug development.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , Antiviral Agents/chemistry , Enterovirus D, Human/enzymology , Evolution, Chemical , 3C Viral Proteases/metabolism , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Biocatalysis , Carbon/chemistry , Enterovirus D, Human/drug effects , Humans , Kinetics , Thermodynamics
9.
Virol Sin ; 35(4): 445-454, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32103448

ABSTRACT

Human rhinoviruses (HRVs) are the predominant infectious agents for the common cold worldwide. The HRV-C species cause severe illnesses in children and are closely related to acute exacerbations of asthma. 3C protease, a highly conserved enzyme, cleaves the viral polyprotein during replication and assists the virus in escaping the host immune system. These key roles make 3C protease an important drug target. A few structures of 3Cs complexed with an irreversible inhibitor rupintrivir have been determined. These structures shed light on the determinants of drug specificity. Here we describe the structures of HRV-C15 3C in free and inhibitor-bound forms. The volume-decreased S1' subsite and half-closed S2 subsite, which were thought to be unique features of enterovirus A 3C proteases, appear in the HRV-C 3C protease. Rupintrivir assumes an "intermediate" conformation in the complex, which might open up additional avenues for the design of potent antiviral inhibitors. Analysis of the features of the three-dimensional structures and the amino acid sequences of 3C proteases suggest new applications for existing drugs.


Subject(s)
3C Viral Proteases/antagonists & inhibitors , 3C Viral Proteases/chemistry , Antiviral Agents/chemistry , Drug Design , Enterovirus A, Human/drug effects , Isoxazoles/chemistry , Phenylalanine/analogs & derivatives , Pyrrolidinones/chemistry , Valine/analogs & derivatives , Crystallography, X-Ray , Enterovirus A, Human/enzymology , Isoxazoles/pharmacology , Models, Molecular , Phenylalanine/chemistry , Phenylalanine/pharmacology , Protein Structure, Tertiary , Pyrrolidinones/pharmacology , Sequence Analysis, DNA , Valine/chemistry , Valine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...