Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Comput Biol Med ; 161: 107004, 2023 07.
Article in English | MEDLINE | ID: covidwho-20243025

ABSTRACT

BACKGROUND: Human neutrophil elastase (HNE) is a key driver of systemic and cardiopulmonary inflammation. Recent studies have established the existence of a pathologically active auto-processed form of HNE with reduced binding affinity against small molecule inhibitors. METHOD: AutoDock Vina v1.2.0 and Cresset Forge v10 software were used to develop a 3D-QSAR model for a series of 47 DHPI inhibitors. Molecular Dynamics (MD) simulations were carried out using AMBER v18 to study the structure and dynamics of sc (single-chain HNE) and tcHNE (two-chain HNE). MMPBSA binding free energies of the previously reported clinical candidate BAY 85-8501 and the highly active BAY-8040 were calculated with sc and tcHNE. RESULTS: The DHPI inhibitors occupy the S1 and S2 subsites of scHNE. The robust 3D-QSAR model showed acceptable predictive and descriptive capability with regression coefficient of r2 = 0.995 and cross-validation regression coefficient q2 = 0.579 for the training set. The key descriptors of shape, hydrophobics and electrostatics were mapped to the inhibitory activity. In auto-processed tcHNE, the S1 subsite undergoes widening and disruption. All the DHPI inhibitors docked with the broadened S1'-S2' subsites of tcHNE with lower AutoDock binding affinities. The MMPBSA binding free energy of BAY-8040 with tcHNE reduced in comparison with scHNE while the clinical candidate BAY 85-8501 dissociated during MD. Thus, BAY-8040 may have lower inhibitory activity against tcHNE whereas the clinical candidate BAY 85-8501 is likely to be inactive. CONCLUSION: SAR insights gained from this study will aid the future development of inhibitors active against both forms of HNE.


Subject(s)
Leukocyte Elastase , Pyrimidinones , Humans , Leukocyte Elastase/chemistry , Leukocyte Elastase/metabolism , Sulfones , Molecular Dynamics Simulation , Quantitative Structure-Activity Relationship , Molecular Docking Simulation
2.
Curr Drug Discov Technol ; 20(4): e220223213933, 2023.
Article in English | MEDLINE | ID: covidwho-2288274

ABSTRACT

BACKGROUND: Tuberculosis (TB) is one of the leading causes of death in the post-COVID- 19 era. It has been observed that there is a devastating condition with a 25-30% increase in TB patients. DNA gyrase B isoform has proved its high potential to be a therapeutically effective target for developing newer and safer anti-TB agents. OBJECTIVE: This study aims to identify minimum structural requirements for the optimization of thiazolopyridine derivatives having DNA gyrase inhibitory activities. Moreover, developed QSAR models could be used to design new thiazolopyridine derivatives and predict their DNA gyrase B inhibitory activity before synthesis. METHODS: 3D-QSAR and Group-based QSAR (G-QSAR) methodologies were adopted to develop accurate, reliable, and predictive QSAR models. Statistical methods such as kNN-MFA SW-FB and MLR SW-FB were used to correlate dependent parameters with descriptors. Both models were thoroughly validated for internal and external predictive abilities. RESULTS: The 3D-QSAR model significantly correlated steric and electrostatic descriptors with q2 0.7491 and predicted r2 0.7792. The G-QSAR model showed that parameters such as SsOHE-index, slogP, ChiV5chain, and T_C_C_3 were crucial for optimizing thiazolopyridine derivatives as DNA gyrase inhibitors. The 3D-QSAR model was interpreted extensively with respect to 3D field points, and the pattern of fragmentation was studied in the G-QSAR model. CONCLUSION: The 3D-QSAR and G-QSAR models were found to be highly predictive. These models could be useful for designing potent DNA gyrase B inhibitors before their synthesis.


Subject(s)
COVID-19 , Tuberculosis , Humans , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Topoisomerase II Inhibitors/chemistry , DNA Gyrase/metabolism , Antitubercular Agents/pharmacology , Quantitative Structure-Activity Relationship
3.
J Comput Chem ; 44(10): 1016-1030, 2023 04 15.
Article in English | MEDLINE | ID: covidwho-2274450

ABSTRACT

Efficacy and safety are among the most desirable characteristics of an ideal drug. The tremendous increase in computing power and the entry of artificial intelligence into the field of computational drug design are accelerating the process of identifying, developing, and optimizing potential drugs. Here, we present novel approach to design new molecules with desired properties. We combined various neural networks and linear regression algorithms to build models for cytotoxicity and anti-HIV activity based on Continual Molecular Interior analysis (CoMIn) and Cinderella's Shoe (CiS) derived molecular descriptors. After validating the reliability of the models, a genetic algorithm was coupled with the Des-Pot Grid algorithm to generate new molecules from a predefined pool of molecular fragments and predict their bioactivity and cytotoxicity. This combination led to the proposal of 16 hit molecules with high anti-HIV activity and low cytotoxicity. The anti-SARS-CoV-2 activity of the hits was predicted.


Subject(s)
Artificial Intelligence , COVID-19 , Humans , Reproducibility of Results , Quantitative Structure-Activity Relationship , Algorithms , Molecular Docking Simulation
4.
Mol Inform ; 42(4): e2200198, 2023 04.
Article in English | MEDLINE | ID: covidwho-2242128

ABSTRACT

The main protease (Mpro ) is an essential enzyme for the life cycle of SARS-CoV-2 and a validated target for treatment of COVID-19 infection. Structure-based pharmacophore modeling combined with QSAR calculations were employed to identify new chemical scaffolds of Mpro inhibitors from natural products repository. Hundreds of pharmacophore models were manually built from their corresponding X-ray crystallographic structures. A pharmacophore model that was validated by receiver operating characteristic (ROC) curve analysis and selected using the statistically optimum QSAR equation was implemented as a 3D-search tool to mine AnalytiCon Discovery database of natural products. Captured hits that showed the highest predicted inhibitory activities were bioassayed. Three active Mpro inhibitors (pseurotin A, lactupicrin, and alpinetin) were successfully identified with IC50 values in low micromolar range.


Subject(s)
Biological Products , COVID-19 , Humans , Models, Molecular , Pharmacophore , Quantitative Structure-Activity Relationship , SARS-CoV-2
5.
J Med Chem ; 66(4): 2744-2760, 2023 02 23.
Article in English | MEDLINE | ID: covidwho-2242001

ABSTRACT

Enveloped viruses depend on the host endoplasmic reticulum (ER) quality control (QC) machinery for proper glycoprotein folding. The endoplasmic reticulum quality control (ERQC) enzyme α-glucosidase I (α-GluI) is an attractive target for developing broad-spectrum antivirals. We synthesized 28 inhibitors designed to interact with all four subsites of the α-GluI active site. These inhibitors are derivatives of the iminosugars 1-deoxynojirimycin (1-DNJ) and valiolamine. Crystal structures of ER α-GluI bound to 25 1-DNJ and three valiolamine derivatives revealed the basis for inhibitory potency. We established the structure-activity relationship (SAR) and used the Site Identification by Ligand Competitive Saturation (SILCS) method to develop a model for predicting α-GluI inhibition. We screened the compounds against SARS-CoV-2 in vitro to identify those with greater antiviral activity than the benchmark α-glucosidase inhibitor UV-4. These host-targeting compounds are candidates for investigation in animal models of SARS-CoV-2 and for testing against other viruses that rely on ERQC for correct glycoprotein folding.


Subject(s)
1-Deoxynojirimycin , Antiviral Agents , COVID-19 , Glycoside Hydrolase Inhibitors , alpha-Glucosidases , Animals , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/pharmacology , alpha-Glucosidases/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Endoplasmic Reticulum/enzymology , Glycoproteins , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , SARS-CoV-2/metabolism , Quantitative Structure-Activity Relationship
6.
Bioorg Med Chem ; 70: 116939, 2022 09 15.
Article in English | MEDLINE | ID: covidwho-2176835
7.
J Microbiol ; 60(12): 1201-1207, 2022 Dec.
Article in English | MEDLINE | ID: covidwho-2117324

ABSTRACT

Candida species cause the most prevalent fungal illness, candidiasis. Candida albicans is known to cause bloodstream infections. This species is a commensal bacterium, but it can cause hospital-acquired diseases, particularly in COVID-19 patients with impaired immune systems. Candida infections have increased in patients with acute respiratory distress syndrome. Coumarins are both naturally occurring and synthetically produced. In this study, the biological activity of 40 coumarin derivatives was used to create a three-dimensional quantitative structure activity relationship (3D-QSAR) model. The training and test minimum inhibitory concentration values of C. albicans active compounds were split, and a regression model based on statistical data was established. This model served as a foundation for the creation of coumarin derivative QSARs. This is a unique way to create new therapeutic compounds for various ailments. We constructed novel structural coumarin derivatives using the derived QSAR model, and the models were confirmed using molecular docking and molecular dynamics simulation.


Subject(s)
COVID-19 , Candidiasis , Humans , Candida albicans , Molecular Docking Simulation , Coumarins/pharmacology , Coumarins/chemistry , Quantitative Structure-Activity Relationship , Antifungal Agents/pharmacology , Antifungal Agents/chemistry
8.
SAR QSAR Environ Res ; 33(10): 753-778, 2022 Oct.
Article in English | MEDLINE | ID: covidwho-2096975

ABSTRACT

Since interleukin-8 (IL-8/CXCL8) and its receptor, CXCR1 and CXCR2, were known in the early 1990s, biological pathways related to these proteins were proven to have high clinical value in cancer and inflammatory/autoimmune conditions treatment. Recently, IL-8 has been identified as biomarker for severe COVID-19 patients and COVID-19 prognosis. Boyles et al. (mAbs 12 (2020), pp. 1831880) have published a high-resolution X-ray crystal structure of the LY3041658 Fab in a complex human CXCL8. They described the ability to bind to IL-8 and the blocking of IL-8/its receptors interaction by the LY3041658 monoclonal antibody. Therefore, the study has been designed to identify potential small molecules inhibiting interleukin-8 by targeting LY3041658/IL-8 complex structure using an in silico approach. A structure­based pharmacophore and molecular docking models of the protein active site cavity were generated to identify possible candidates, followed by virtual screening with the ZINC database. ADME analysis of hit compounds was also conducted. Molecular dynamics simulations were then performed to survey the behaviour and stability of the ligand-protein complexes. Furthermore, the MM/PBSA technique has been utilized to evaluate the free binding energy. The final data confirmed that one newly obtained compound, ZINC21882765, may serve as the best potential inhibitor for IL-8.


Subject(s)
COVID-19 Drug Treatment , Interleukin-8 , Humans , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Molecular Dynamics Simulation , Ligands
9.
Int J Mol Sci ; 23(18)2022 Sep 19.
Article in English | MEDLINE | ID: covidwho-2071502

ABSTRACT

The chymotrypsin-like cysteine protease (3CLpro, also known as main protease-Mpro) and papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been used as the main targets for screening potential synthetic inhibitors for posterior in vitro evaluation of the most promising compounds. In this sense, the present work reports for the first time the evaluation of the interaction between Mpro/PLpro with a series of 17 porphyrin analogues-corrole (C1), meso-aryl-corrole (C2), and 15 fluorinated-meso-aryl-corrole derivatives (C3-C17) via molecular docking calculations. The impact of fluorine atoms on meso-aryl-corrole structure was also evaluated in terms of binding affinity and physical-chemical properties by two-dimensional quantitative structure-activity relationship (2D-QSAR). The presence of phenyl moieties increased the binding capacity of corrole for both proteases and depending on the position of fluorine atoms might impact positively or negatively the binding capacity. For Mpro the para-fluorine atoms might decrease drastically the binding capacity, while for PLpro there was a certain increase in the binding affinity of fluorinated-corroles with the increase of fluorine atoms into meso-aryl-corrole structure mainly from tri-fluorinated insertions. The 2D-QSAR models indicated two separated regions of higher and lower affinity for Mpro:C1-C17 based on dual electronic parameters (σI and σR), as well as one model was obtained with a correlation between the docking score value of Mpro:C2-C17 and the corresponding 13C nuclear magnetic resonance (NMR) chemical shifts of the sp2 carbon atoms (δC-1 and δC-2) of C2-C17. Overall, the fluorinated-meso-aryl-corrole derivatives showed favorable in silico parameters as potential synthetic compounds for future in vitro assays on the inhibition of SARS-CoV-2 replication.


Subject(s)
COVID-19 Drug Treatment , Porphyrins , Antiviral Agents/pharmacology , Carbon , Chymotrypsin , Coronavirus 3C Proteases , Fluorine , Humans , Molecular Docking Simulation , Papain , Peptide Hydrolases , Porphyrins/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Quantitative Structure-Activity Relationship , SARS-CoV-2
10.
Future Med Chem ; 14(21): 1541-1559, 2022 11.
Article in English | MEDLINE | ID: covidwho-2055773

ABSTRACT

Background: In the recent COVID-19 pandemic, SARS-CoV-2 infection spread worldwide. The 3C-like protease (3CLpro) is a promising drug target for SARS-CoV-2. Results: We constructed a deep learning-based convolutional neural network-quantitative structure-activity relationship (CNN-QSAR) model and deployed it on various databases to predict the biological activity of 3CLpro inhibitors. Subsequently, molecular docking analysis, molecular dynamics simulations and binding free energy calculations were performed to validate the predicted inhibitory activity against 3CLpro of SARS-CoV-2. The model showed mean squared error = 0.114, mean absolute error = 0.24 and predicted R2 = 0.84 for the test dataset. Diosmin showed good binding affinity and stability over the course of the simulations. Conclusion: The results suggest that the proposed CNN-QSAR model can be an efficient method for hit prediction and a new way to identify hit compounds against 3CLpro of SARS-CoV-2.


Subject(s)
COVID-19 , Deep Learning , Humans , SARS-CoV-2 , Quantitative Structure-Activity Relationship , Coronavirus 3C Proteases , Pandemics , Molecular Docking Simulation , Peptide Hydrolases , Protease Inhibitors/chemistry , Molecular Dynamics Simulation , Antiviral Agents/pharmacology
11.
SAR QSAR Environ Res ; 33(9): 649-675, 2022 Sep.
Article in English | MEDLINE | ID: covidwho-2008373

ABSTRACT

The pandemic of COVID-19 caused by SARS-CoV-2 has made a worldwide health emergency. Despite the fact that current vaccines are readily available, several SARSCoV-2 variants affecting the existing vaccine are to be less effective due to the mutations in the structural proteins. Furthermore, the appearance of the new variants cannot be easily predicted in the future. Therefore, the attempts to construct new vaccines or to modify the current vaccines are still pivotal works for preventing the spread of the virus. In the present investigation, the computational analysis through immunoinformatics, molecular docking, and molecular dynamics (MD) simulation is employed to construct an effective vaccine against SARS-CoV2. The structural proteins of SARS-CoV2 are utilized to create a multiepitope-based vaccine (MEV). According to our findings presented by systematic procedures in the current investigation, the MEV construct may be able to trigger a strong immunological response against the virus. Therefore, the designed MEV could be a potential vaccine candidate against SARS-CoV-2, and also it is expected to be effective for other variants.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/prevention & control , COVID-19 Vaccines , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Humans , Immunogenicity, Vaccine , Molecular Docking Simulation , Molecular Dynamics Simulation , Quantitative Structure-Activity Relationship , RNA, Viral , Vaccines, Subunit/chemistry
12.
Int J Mol Sci ; 23(3)2022 Jan 30.
Article in English | MEDLINE | ID: covidwho-1917507

ABSTRACT

Traditionally, drug development involved the individual synthesis and biological evaluation of hundreds to thousands of compounds with the intention of highlighting their biological activity, selectivity, and bioavailability, as well as their low toxicity. On average, this process of new drug development involved, in addition to high economic costs, a period of several years before hopefully finding a drug with suitable characteristics to drive its commercialization. Therefore, the chemical synthesis of new compounds became the limiting step in the process of searching for or optimizing leads for new drug development. This need for large chemical libraries led to the birth of high-throughput synthesis methods and combinatorial chemistry. Virtual combinatorial chemistry is based on the same principle as real chemistry-many different compounds can be generated from a few building blocks at once. The difference lies in its speed, as millions of compounds can be produced in a few seconds. On the other hand, many virtual screening methods, such as QSAR (Quantitative Sturcture-Activity Relationship), pharmacophore models, and molecular docking, have been developed to study these libraries. These models allow for the selection of molecules to be synthesized and tested with a high probability of success. The virtual combinatorial chemistry-virtual screening tandem has become a fundamental tool in the process of searching for and developing a drug, as it allows the process to be accelerated with extraordinary economic savings.


Subject(s)
Combinatorial Chemistry Techniques/methods , Small Molecule Libraries/pharmacology , Drug Design , Models, Molecular , Molecular Docking Simulation , Quantitative Structure-Activity Relationship
13.
J Comput Aided Mol Des ; 36(7): 483-505, 2022 07.
Article in English | MEDLINE | ID: covidwho-1899232

ABSTRACT

The main protease (Mpro) of SARS-Cov-2 is the essential enzyme for maturation of functional proteins implicated in viral replication and transcription. The peculiarity of its specific cleavage site joint with its high degree of conservation among all coronaviruses promote it as an attractive target to develop broad-spectrum inhibitors, with high selectivity and tolerable safety profile. Herein is reported a combination of three-dimensional quantitative structure-activity relationships (3-D QSAR) and comparative molecular binding energy (COMBINE) analysis to build robust and predictive ligand-based and structure-based statistical models, respectively. Models were trained on experimental binding poses of co-crystallized Mpro-inhibitors and validated on available literature data. By means of deep optimization both models' goodness and robustness reached final statistical values of r2/q2 values of 0.97/0.79 and 0.93/0.79 for the 3-D QSAR and COMBINE approaches respectively, and an overall predictiveness values of 0.68 and 0.57 for the SDEPPRED and AAEP metrics after application to a test set of 60 compounds covered by the training set applicability domain. Despite the different nature (ligand-based and structure-based) of the employed methods, their outcome fully converged. Furthermore, joint ligand- and structure-based structure-activity relationships were found in good agreement with nirmatrelvir chemical features properties, a novel oral Mpro-inhibitor that has recently received U.S. FDA emergency use authorization (EUA) for the oral treatment of mild-to-moderate COVID-19 infected patients. The obtained results will guide future rational design and/or virtual screening campaigns with the aim of discovering new potential anti-coronavirus lead candidates, minimizing both time and financial resources. Moreover, as most of calculation were performed through the well-established web portal 3d-qsar.com the results confirm the portal as a useful tool for drug design.


Subject(s)
COVID-19 Drug Treatment , Quantitative Structure-Activity Relationship , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Coronavirus 3C Proteases , Humans , Ligands , Molecular Docking Simulation , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , SARS-CoV-2
14.
SAR QSAR Environ Res ; 33(5): 341-356, 2022 May.
Article in English | MEDLINE | ID: covidwho-1819655

ABSTRACT

The outbreak of coronavirus disease 2019 (COVID-19) at the end of 2019 affected global health. Its infection agent was called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Wearing a mask, maintaining social distance, and vaccination are effective ways to prevent infection of SARS-CoV-2, but none of them help infected people. Targeting the enzymes of SARS-CoV-2 is an effective way to stop the replication of the virus in infected people and treat COVID-19 patients. SARS-CoV-2 main protease is a therapeutic target which the inhibition of its enzymatic activity prevents from the replication of SARS-CoV-2. A large database of molecules has been searched to identify new inhibitors for SARS-CoV-2 main protease enzyme. At the first step, ligand screening based on similarity search was used to select similar compounds to known SARS-CoV-2 main protease inhibitors. Then molecules with better predicted pharmacokinetic properties were selected. Structure-based virtual screening based on the application of molecular docking and molecular dynamics simulation methods was used to select more effective inhibitors among selected molecules in previous step. Finally two compounds were considered as SARS-CoV-2 main protease inhibitors.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Antiviral Agents/pharmacology , Computers , Coronavirus 3C Proteases , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , Quantitative Structure-Activity Relationship
15.
SAR QSAR Environ Res ; 33(5): 357-386, 2022 May.
Article in English | MEDLINE | ID: covidwho-1774080

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) outbreak is posing a serious public health threat worldwide in the form of COVD-19. Herein, we have performed two-dimensional quantitative structure-activity relationship (2D-QSAR) and three-dimensional pharmacophore modelling analysis employing inhibitors of 3-chymotrypsin-like protease (3CLpro), the leading protease that is crucial for the replication of SARS-CoV-2. The investigation aims to identify the important structural features responsible for the enzyme inhibition and the search for novel 3CLpro enzyme inhibitors as effective therapeutics for treating SARS-CoV-2. Furthermore, we carried out molecular docking studies using the most and least active compounds in the dataset, aiming to validate the contributions of various features as appeared in the QSAR models. Later, the stringently validated 2D-QSAR model was used to estimate the 3CLpro inhibitory activity of compounds from five chemical databases. Compounds with the significant predicted activity were then subjected to pharmacophore-based virtual screening to screen the top-rated compounds, which were then further subjected to molecular docking analysis, absorption, distribution, metabolism, excretion - toxicity (ADMET) profiling, and molecular dynamics (MD) simulation. The multi-step virtual screening analyses suggested that compounds CASAntiV-865453-58-3, CASAntiV-865453-40-3, and CASAntiV-2043031-84-9 could be used as effective therapeutic agents for the treatment of SARS-CoV-2.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Quantitative Structure-Activity Relationship
16.
Med Chem ; 18(8): 871-883, 2022.
Article in English | MEDLINE | ID: covidwho-1631502

ABSTRACT

BACKGROUND: Chemokines are involved in several human diseases and different stages of COVID-19 infection. They play a critical role in the pathophysiology of the associated acute respiratory disease syndrome, a major complication leading to death among COVID-19 patients. In particular, CXC chemokine receptor 4 (CXCR4) was found to be highly expressed in COVID-19 patients. METHODS: We herein describe a computational workflow based on combining pharmacophore modeling and QSAR analysis towards the discovery of novel CXCR4 inhibitors. Subsequent virtual screening identified two promising CXCR4 inhibitors from the National Cancer Institute (NCI) list of compounds. The most active hit showed in vitro IC50 value of 24.4 µM. CONCLUSION: These results proved the validity of the QSAR model and associated pharmacophore models as means to screen virtual databases for new CXCR4 inhibitors as leads for the development of new COVID-19 therapies.


Subject(s)
COVID-19 Drug Treatment , Quantitative Structure-Activity Relationship , Receptors, CXCR4 , Humans , Ligands , Molecular Docking Simulation , Receptors, CXCR4/antagonists & inhibitors
17.
SAR QSAR Environ Res ; 32(11): 863-888, 2021 Nov.
Article in English | MEDLINE | ID: covidwho-1606722

ABSTRACT

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.


Subject(s)
Antiviral Agents/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Protease Inhibitors/chemistry , SARS-CoV-2/drug effects , Antiviral Agents/pharmacokinetics , Coronavirus 3C Proteases/chemistry , Inhibitory Concentration 50 , Molecular Docking Simulation , Neural Networks, Computer , Protease Inhibitors/pharmacokinetics , Quantitative Structure-Activity Relationship , Reproducibility of Results , SARS-CoV-2/enzymology
18.
Biomolecules ; 11(12)2021 12 04.
Article in English | MEDLINE | ID: covidwho-1554985

ABSTRACT

Inflammation involves a complex biological response of the body tissues to damaging stimuli. When dysregulated, inflammation led by biomolecular mediators such as caspase-1 and tumor necrosis factor-alpha (TNF-alpha) can play a detrimental role in the progression of different medical conditions such as cancer, neurological disorders, autoimmune diseases, and cytokine storms caused by viral infections such as COVID-19. Computational approaches can accelerate the search for dual-target drugs able to simultaneously inhibit the aforementioned proteins, enabling the discovery of wide-spectrum anti-inflammatory agents. This work reports the first multicondition model based on quantitative structure-activity relationships and a multilayer perceptron neural network (mtc-QSAR-MLP) for the virtual screening of agency-regulated chemicals as versatile anti-inflammatory therapeutics. The mtc-QSAR-MLP model displayed accuracy higher than 88%, and was interpreted from a physicochemical and structural point of view. When using the mtc-QSAR-MLP model as a virtual screening tool, we could identify several agency-regulated chemicals as dual inhibitors of caspase-1 and TNF-alpha, and the experimental information later retrieved from the scientific literature converged with our computational results. This study supports the capabilities of our mtc-QSAR-MLP model in anti-inflammatory therapy with direct applications to current health issues such as the COVID-19 pandemic.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Caspase Inhibitors/pharmacology , Drug Repositioning/methods , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Anti-Inflammatory Agents/chemistry , Caspase 1/metabolism , Caspase Inhibitors/chemistry , Humans , Inflammation/drug therapy , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , COVID-19 Drug Treatment
19.
SAR QSAR Environ Res ; 32(12): 963-983, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1532255

ABSTRACT

The coronavirus helicase is an essential enzyme required for viral replication/transcription pathways. Structural studies revealed a sulphate moiety that interacts with key residues within the nucleotide-binding site of the helicase. Compounds with a sulphoxide or a sulphone moiety could interfere with these interactions and consequently inhibit the enzyme. The molecular operating environment (MOE) was used to dock 189 sulphoxide and sulphone-containing FDA-approved compounds to the nucleotide-binding site. Zafirlukast, a leukotriene receptor antagonist used to treat chronic asthma, achieved the lowest docking score at -8.75 kcals/mol. The inhibitory effect of the compounds on the SARS-CoV-2 helicase dsDNA unwinding activity was tested by a FRET-based assay. Zafirlukast was the only compound to inhibit the enzyme (IC50 = 16.3 µM). The treatment of Vero E6 cells with 25 µM zafirlukast prior to SARS-CoV-2 infection decreased the cytopathic effects of SARS-CoV-2 significantly. These results suggest that zafirlukast alleviates SARS-CoV-2 pathogenicity by inhibiting the viral helicase and impairing the viral replication/transcription pathway. Zafirlukast could be clinically developed as a new antiviral treatment for SARS-CoV-2 and other coronavirus diseases. This discovery is based on molecular modelling, in vitro inhibition of the SARS-CoV helicase activity and cell-based SARS-CoV-2 viral replication.


Subject(s)
Antiviral Agents/pharmacology , DNA Helicases/antagonists & inhibitors , Indoles/pharmacology , Phenylcarbamates/pharmacology , SARS-CoV-2/drug effects , Sulfonamides/pharmacology , Animals , Chlorocebus aethiops , Fluorescence Resonance Energy Transfer , Quantitative Structure-Activity Relationship , SARS-CoV-2/enzymology , Vero Cells , Virus Replication/drug effects , COVID-19 Drug Treatment
20.
ChemMedChem ; 16(22): 3418-3427, 2021 11 19.
Article in English | MEDLINE | ID: covidwho-1525425

ABSTRACT

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).


Subject(s)
Antiviral Agents/pharmacology , Pyrazines/pharmacology , SARS-CoV-2/drug effects , Amides/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Antiviral Agents/toxicity , Chlorocebus aethiops , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Pyrazines/chemical synthesis , Pyrazines/metabolism , Pyrazines/toxicity , Quantitative Structure-Activity Relationship , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL