1
|
Sheng YJ, Kuo STA, Yang T, Russell DH, Yan X, Xu S, Liu WR, Fierke CA. BRD4354 Is a Potent Covalent Inhibitor against the SARS-CoV-2 Main Protease. Biochemistry 2024:10.1021/acs.biochem.3c00685. [PMID: 38329238 PMCID: PMC11306412 DOI: 10.1021/acs.biochem.3c00685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Numerous organic molecules are known to inhibit the main protease (MPro) of SARS-CoV-2, the pathogen of Coronavirus Disease 2019 (COVID-19). Guided by previous research on zinc-ligand inhibitors of MPro and zinc-dependent histone deacetylases (HDACs), we identified BRD4354 as a potent inhibitor of MPro. The in vitro protease activity assays show that BRD4354 displays time-dependent inhibition against MPro with an IC50 (concentration that inhibits activity by 50%) of 0.72 ± 0.04 μM after 60 min of incubation. Inactivation follows a two-step process with an initial rapid binding step with a KI of 1.9 ± 0.5 μM followed by a second slow inactivation step, kinact,max of 0.040 ± 0.002 min-1. Native mass spectrometry studies indicate that a covalent intermediate is formed where the ortho-quinone methide fragment of BRD4354 forms a covalent bond with the catalytic cysteine C145 of MPro. Based on these data, a Michael-addition reaction mechanism between MPro C145 and BRD4354 was proposed. These results suggest that both preclinical testing of BRD4354 and structure-activity relationship studies based on BRD4354 are warranted to develop more effective anti-COVID therapeutics.
Collapse
Affiliation(s)
- Yan J. Sheng
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Syuan-Ting Alex Kuo
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Tingyuan Yang
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - David H. Russell
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Xin Yan
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Shiqing Xu
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
| | - Wenshe R. Liu
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
- Institute of Biosciences and Technology and Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX 77030, USA
- Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Carol A. Fierke
- Department of Biochemistry, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
2
|
Karakkadparambil Sankaran S, Nair AS. Molecular dynamics and docking studies on potentially active natural phytochemicals for targeting SARS-CoV-2 main protease. J Biomol Struct Dyn 2022:1-17. [PMID: 35930306 DOI: 10.1080/07391102.2022.2107573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
In the present study, we screened eighty seven novel phytochemical compounds from four popular herbs, such as, Aegle Marmelos, Coleus Amboinicus, Aerva Lanata and Biophytum Sensitivum and identified the best three for targeting the main protease (Mpro) receptor of SARS-CoV-2. After categorizing all the phytochemicals based upon LibDock scores and hydrogen bonding interactions, the top ranked 26 compounds were further subjected for detailed Molecular dynamics (MD) study. From these screening we identified that Aegelinosides B leads the list with a high LibDock value of 142.50 (binding energy: -8.54 kcal/mol), which is better than several popular reference compounds namely, Tipranavir (LibDock score, 141.50), Saquinavir (125.34), Zopicole (122.9), Pirenepine (122.70), (115.37), Metixene (109.18), Oxiconazole Pimozide (138.00) and Rimonabant (91.88). Detailed analysis for structural stability (RMSD), Cα fluctuations (RMSF), intermolecular hydrogen bond interactions, effect of solvent accessibility (SASA) and compactness (Rg) factors were performed for the best six compounds and it is found that they are very stable and exhibit folding behavior. Apart from the docking and MD tests, through further drug-likeness and toxicity tests, three compounds, such as, Aegelinosides B, Epicatechin, and Feruloyltyramine (LibDock scores, respectively, 142.50, 124.33 and 129.06) can be suggested for fighting SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Achuthsankar S Nair
- Department of Computational Biology and Bioinformatics, University of Kerala, Thiruvananthapuram, Kerala, India
| |
Collapse
|
3
|
Proia E, Ragno A, Antonini L, Sabatino M, Mladenovič M, Capobianco R, Ragno R. Ligand-based and structure-based studies to develop predictive models for SARS-CoV-2 main protease inhibitors through the 3d-qsar.com portal. J Comput Aided Mol Des 2022; 36:483-505. [PMID: 35716228 PMCID: PMC9206107 DOI: 10.1007/s10822-022-00460-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/28/2022] [Indexed: 11/05/2022]
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.
Collapse
Affiliation(s)
- Eleonora Proia
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Alessio Ragno
- Department of Computer, Control and Management Engineering "Antonio Ruberti", Sapienza University of Rome, Rome, Italy
| | - Lorenzo Antonini
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Manuela Sabatino
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Milan Mladenovič
- Department of Chemistry, Faculty of Science, Kragujevac Center for Computational Biochemistry, University of Kragujevac, Radoja Domanovića 12, P.O. Box 60, 34000, Kragujevac, Serbia
| | - Roberto Capobianco
- Department of Computer, Control and Management Engineering "Antonio Ruberti", Sapienza University of Rome, Rome, Italy
- Sony AI, Rome, Italy
| | - Rino Ragno
- Department of Drug Chemistry and Technology, Rome Center for Molecular Design, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
4
|
Shih PC, Mao YW, Hu JW, Hsieh HY, Shih TM, Lu LP, Chang WH, Huang CH, Lin CH, Lin CH, Tan P, Yang YC, Chien MH, Su CC, Yeh CH, Chuang PY, Hsieh TL, Wang CC, Hsieh PS, Chou TY, Tsai GE. Development of Ultrapure and Potent Tannic Acids as a Pan-coronal Antiviral Therapeutic. ACS Pharmacol Transl Sci 2022; 5:400-412. [PMID: 37582235 PMCID: PMC9128009 DOI: 10.1021/acsptsci.1c00264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The rampageous transmission of SARS-CoV-2 has been devastatingly impacting human life and public health since late 2019. The waves of pandemic events caused by distinct coronaviruses at present and over the past decades have prompted the need to develop broad-spectrum antiviral drugs against them. In this study, our Pentarlandir ultrapure and potent tannic acids (UPPTA) showed activities against two coronaviral strains, SARS-CoV-2 and HCoV-OC43, the earliest-known coronaviruses. The mode of inhibition of Pentarlandir UPPTA is likely to act on 3-chymotrypsin-like protease (3CLpro) to prevent viral replication, as supported by results of biochemical analysis, a 3CLpro assay, and a "gain-of-function" 3CLpro overexpressed cell-based method. Even in the 3CLpro overexpressed environment, Pentarlandir UPPTA remained its antiviral characteristic. Utilizing cell-based virucidal and cytotoxicity assays, the 50% effective concentrations (EC50) and 50% cytotoxicity concentration (CC50) of Pentarlandir UPPTA were determined to be ∼0.5 and 52.5 μM against SARS-CoV-2, while they were 1.3 and 205.9 μM against HCoV-OC43, respectively. In the pharmacokinetic studies, Pentarlandir UPPTA was distributable at a high level to the lung tissue with no accumulation in the body, although the distribution was affected by the food effect. With further investigation in toxicology, Pentarlandir UPPTA demonstrated an overall safe toxicology profile. Taking these findings together, Pentarlandir UPPTA is considered to be a safe and efficacious pancoronal antiviral drug candidate that has been advanced to clinical development.
Collapse
Affiliation(s)
- Po-Chang Shih
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Yi-Wen Mao
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Jhe-Wei Hu
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Han-Yi Hsieh
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Tsai-Miao Shih
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Lu-Ping Lu
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Wei-Hua Chang
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Chan-Hui Huang
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Chia-Hung Lin
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Chih-Hung Lin
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Peng Tan
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Ya-Ching Yang
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Ming-Hong Chien
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Chen-Che Su
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Cheng-Hsin Yeh
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Pei-Yun Chuang
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Tien-Lan Hsieh
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Ching-Cheng Wang
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
| | - Po-Shiuan Hsieh
- Institute
of Physiology, National Defense Medical
Center, Taipei
City 11490, Taiwan
- Department
of Medical Research, Tri-Service General
Hospital, Taipei City 114, Taiwan
| | - Teh-Ying Chou
- Department
of Pathology and Laboratory Medicine, Taipei
Veterans General Hospital, Taipei City 112, Taiwan
- Institute
of Clinical Medicine, National Yang-Ming
Chiao Tung University, Taipei City 112304, Taiwan
| | - Guochuan Emil Tsai
- Department
of Research and Development, SyneuRx International
(Taiwan) Corp., New Taipei City 22175, Taiwan
- UCLA School
of Medicine, Los Angeles, California 90095, United States
| |
Collapse
|
5
|
Antonopoulou I, Sapountzaki E, Rova U, Christakopoulos P. Inhibition of the main protease of SARS-CoV-2 (M pro) by repurposing/designing drug-like substances and utilizing nature's toolbox of bioactive compounds. Comput Struct Biotechnol J 2022; 20:1306-1344. [PMID: 35308802 PMCID: PMC8920478 DOI: 10.1016/j.csbj.2022.03.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 12/14/2022] Open
Abstract
The emergence of the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has resulted in a long pandemic, with numerous cases and victims worldwide and enormous consequences on social and economic life. Although vaccinations have proceeded and provide a valuable shield against the virus, the approved drugs are limited and it is crucial that further ways to combat infection are developed, that can also act against potential mutations. The main protease (Mpro) of the virus is an appealing target for the development of inhibitors, due to its importance in the viral life cycle and its high conservation among different coronaviruses. Several compounds have shown inhibitory potential against Mpro, both in silico and in vitro, with few of them also having entered clinical trials. These candidates include: known drugs that have been repurposed, molecules specifically designed based on the natural substrate of the protease or on structural moieties that have shown high binding affinity to the protease active site, as well as naturally derived compounds, either isolated or in plant extracts. The aim of this work is to collectively present the results of research regarding Mpro inhibitors to date, focusing on the function of the compounds founded by in silico simulations and further explored by in vitro and in vivo assays. Creating an extended portfolio of promising compounds that may block viral replication by inhibiting Mpro and by understanding involved structure-activity relationships, could provide a basis for the development of effective solutions against SARS-CoV-2 and future related outbreaks.
Collapse
Affiliation(s)
| | | | - Ulrika Rova
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental and Natural Resources Engineering, Luleå University of Technology, SE-97187 Luleå, Sweden
| | - Paul Christakopoulos
- Biochemical Process Engineering, Division of Chemical Engineering, Department of Civil, Environmental and Natural Resources Engineering, Luleå University of Technology, SE-97187 Luleå, Sweden
| |
Collapse
|
6
|
Evaluation of COVID-19 protease and HIV inhibitors interactions. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2022; 72:1-8. [PMID: 36651526 DOI: 10.2478/acph-2022-0010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/27/2021] [Indexed: 01/20/2023]
Abstract
The epidemic of the novel coronavirus disease (COVID-19) that started in 2019 has evoked an urgent demand for finding new potential therapeutic agents. In this study, we performed a molecular docking of anti-HIV drugs to refine HIV protease inhibitors and nucleotide analogues to target COVID-19. The evaluation was based on docking scores calculated by AutoDock Vina and top binding poses were analyzed. Our results suggested that lopinavir, darunavir, atazanavir, remdesivir, and tipranavir have the best binding affinity for the 3-chymotrypsin-like protease of COVID-19. The comparison of the binding sites of three drugs, namely, darunavir, atazanavir and remdesivir, showed an overlap region of the protein pocket. Our study showed a strong affinity between lopinavir, darunavir, atazanavir, tipranavir and COVID-19 protease. However, their efficacy should be confirmed by in vitro studies since there are concerns related to interference with their active sites.
Collapse
|
7
|
Kevill JL, Pellett C, Farkas K, Brown MR, Bassano I, Denise H, McDonald JE, Malham SK, Porter J, Warren J, Evens NP, Paterson S, Singer AC, Jones DL. A comparison of precipitation and filtration-based SARS-CoV-2 recovery methods and the influence of temperature, turbidity, and surfactant load in urban wastewater. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 808:151916. [PMID: 34826466 PMCID: PMC8610557 DOI: 10.1016/j.scitotenv.2021.151916] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 05/09/2023]
Abstract
Wastewater-based epidemiology (WBE) has become a complimentary surveillance tool during the SARS-CoV-2 pandemic. Viral concentration methods from wastewater are still being optimised and compared, whilst viral recovery under different wastewater characteristics and storage temperatures remains poorly understood. Using urban wastewater samples, we tested three viral concentration methods; polyethylene glycol precipitation (PEG), ammonium sulphate precipitation (AS), and CP select™ InnovaPrep® (IP) ultrafiltration. We found no major difference in SARS-CoV-2 and faecal indicator virus (crAssphage) recovery from wastewater samples (n = 46) using these methods, PEG slightly (albeit non-significantly), outperformed AS and IP for SARS-CoV-2 detection, as a higher genome copies per litre (gc/l) was recorded for a larger proportion of samples. Next generation sequencing of 8 paired samples revealed non-significant differences in the quality of data between AS and IP, though IP data quality was slightly better and less variable. A controlled experiment assessed the impact of wastewater suspended solids (turbidity; 0-400 NTU), surfactant load (0-200 mg/l), and storage temperature (5-20 °C) on viral recovery using the AS and IP methods. SARS-CoV-2 recoveries were >20% with AS and <10% with IP in turbid samples, whilst viral recoveries for samples with additional surfactant were between 0-18% for AS and 0-5% for IP. Turbidity and sample storage temperature combined had no significant effect on SARS-CoV-2 recovery (p > 0.05), whilst surfactant and storage temperature combined were significant negative correlates (p < 0.001 and p < 0.05, respectively). In conclusion, our results show that choice of methodology had small effect on viral recovery of SARS-CoV-2 and crAssphage in wastewater samples within this study. In contrast, sample turbidity, storage temperature, and surfactant load did affect viral recovery, highlighting the need for careful consideration of the viral concentration methodology used when working with wastewater samples.
Collapse
Affiliation(s)
- Jessica L Kevill
- Centre for Environmental Biotechnology, School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK.
| | - Cameron Pellett
- Centre for Environmental Biotechnology, School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Kata Farkas
- Centre for Environmental Biotechnology, School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK; School of Ocean Sciences, Bangor University, Menai Bridge, Anglesey LL59 5AB, UK
| | - Mathew R Brown
- Joint Biosecurity Centre, Department of Health and Social Care, London WC1B 4DA, UK; School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Irene Bassano
- Joint Biosecurity Centre, Department of Health and Social Care, London WC1B 4DA, UK; Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Hubert Denise
- Joint Biosecurity Centre, Department of Health and Social Care, London WC1B 4DA, UK
| | - James E McDonald
- Centre for Environmental Biotechnology, School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Shelagh K Malham
- School of Ocean Sciences, Bangor University, Menai Bridge, Anglesey LL59 5AB, UK
| | - Jonathan Porter
- Environment Agency National Laboratory Service, Exeter, Devon EX6 8PE, UK
| | - Jonathan Warren
- Environment Agency National Laboratory Service, Exeter, Devon EX6 8PE, UK
| | - Nicholas P Evens
- Environment Agency National Laboratory Service, Exeter, Devon EX6 8PE, UK
| | - Steve Paterson
- Centre of Genomics Research & NERC Environmental Omics Facility, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Davey L Jones
- Centre for Environmental Biotechnology, School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK; Food Futures Institute, Murdoch University, 90 South Street, Murdoch, WA 6105, Australia
| |
Collapse
|
8
|
Fred SM, Kuivanen S, Ugurlu H, Casarotto PC, Levanov L, Saksela K, Vapalahti O, Castrén E. Antidepressant and Antipsychotic Drugs Reduce Viral Infection by SARS-CoV-2 and Fluoxetine Shows Antiviral Activity Against the Novel Variants in vitro. Front Pharmacol 2022; 12:755600. [PMID: 35126106 PMCID: PMC8809408 DOI: 10.3389/fphar.2021.755600] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/09/2021] [Indexed: 12/29/2022] Open
Abstract
Repurposing of currently available drugs is a valuable strategy to tackle the consequences of COVID-19. Recently, several studies have investigated the effect of psychoactive drugs on SARS-CoV-2 in cell culture models as well as in clinical practice. Our aim was to expand these studies and test some of these compounds against newly emerged variants. Several antidepressants and antipsychotic drugs with different primary mechanisms of action were tested in ACE2/TMPRSS2-expressing human embryonic kidney cells against the infection by SARS-CoV-2 spike protein-dependent pseudoviruses. Some of these compounds were also tested in human lung epithelial cell line, Calu-1, against the first wave (B.1) lineage of SARS-CoV-2 and the variants of concern, B.1.1.7, B.1.351, and B.1.617.2. Several clinically used antidepressants, including fluoxetine, citalopram, reboxetine, imipramine, as well as antipsychotic compounds chlorpromazine, flupenthixol, and pimozide inhibited the infection by pseudotyped viruses with minimal effects on cell viability. The antiviral action of several of these drugs was verified in Calu-1 cells against the B.1 lineage of SARS-CoV-2. By contrast, the anticonvulsant carbamazepine, and novel antidepressants ketamine, known as anesthetic at high doses, and its derivatives as well as MAO and phosphodiesterase inhibitors phenelzine and rolipram, respectively, showed no activity in the pseudovirus model. Furthermore, fluoxetine remained effective against pseudoviruses with common receptor binding domain mutations, N501Y, K417N, and E484K, as well as B.1.1.7 (alpha), B.1.351 (beta), and B.1.617.2 (delta) variants of SARS-CoV-2. Our study confirms previous data and extends information on the repurposing of these drugs to counteract SARS-CoV-2 infection including different variants of concern, however, extensive clinical studies must be performed to confirm our in vitro findings.
Collapse
Affiliation(s)
- Senem Merve Fred
- Neuroscience Center–HiLIFE, University of Helsinki, Helsinki, Finland
| | - Suvi Kuivanen
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Hasan Ugurlu
- Department of Virology, University of Helsinki, Helsinki, Finland
| | | | - Lev Levanov
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Kalle Saksela
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center–HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Piplani S, Singh PK, Winkler DA, Petrovsky N. Computationally repurposed drugs and natural products against RNA dependent RNA polymerase as potential COVID-19 therapies. MOLECULAR BIOMEDICINE 2021; 2:28. [PMID: 34766004 PMCID: PMC8450033 DOI: 10.1186/s43556-021-00050-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022] Open
Abstract
Repurposing of existing drugs and drug candidates is an ideal approach to identify new potential therapies for SARS-CoV-2 that can be tested without delay in human trials of infected patients. Here we applied a virtual screening approach using Autodock Vina and molecular dynamics simulation in tandem to calculate binding energies for repurposed drugs against the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp). We thereby identified 80 promising compounds with potential activity against SARS-Cov2, consisting of a mixture of antiviral drugs, natural products and drugs with diverse modes of action. A substantial proportion of the top 80 compounds identified in this study had been shown by others to have SARS-CoV-2 antiviral effects in vitro or in vivo, thereby validating our approach. Amongst our top hits not previously reported to have SARS-CoV-2 activity, were eribulin, a macrocyclic ketone analogue of the marine compound halichondrin B and an anticancer drug, the AXL receptor tyrosine kinase inhibitor bemcentinib. Our top hits from our RdRp drug screen may not only have utility in treating COVID-19 but may provide a useful starting point for therapeutics against other coronaviruses. Hence, our modelling approach successfully identified multiple drugs with potential activity against SARS-CoV-2 RdRp. Supplementary Information The online version contains supplementary material available at 10.1186/s43556-021-00050-3.
Collapse
Affiliation(s)
- Sakshi Piplani
- College of Medicine and Public Health, Flinders University, Bedford Park, 5046 Australia
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, 5046 Australia
| | | | - David A. Winkler
- School of Biochemistry and Genetics, La Trobe University, Bundoora, 3086 Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052 Australia
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD UK
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Bedford Park, 5046 Australia
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, 5046 Australia
| |
Collapse
|
10
|
Llanos MA, Gantner ME, Rodriguez S, Alberca LN, Bellera CL, Talevi A, Gavernet L. Strengths and Weaknesses of Docking Simulations in the SARS-CoV-2 Era: the Main Protease (Mpro) Case Study. J Chem Inf Model 2021; 61:3758-3770. [PMID: 34313128 DOI: 10.1021/acs.jcim.1c00404] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The scientific community is working against the clock to arrive at therapeutic interventions to treat patients with COVID-19. Among the strategies for drug discovery, virtual screening approaches have the capacity to search potential hits within millions of chemical structures in days, with the appropriate computing infrastructure. In this article, we first analyzed the published research targeting the inhibition of the main protease (Mpro), one of the most studied targets of SARS-CoV-2, by docking-based methods. An alarming finding was the lack of an adequate validation of the docking protocols (i.e., pose prediction and virtual screening accuracy) before applying them in virtual screening campaigns. The performance of the docking protocols was tested at some level in 57.7% of the 168 investigations analyzed. However, we found only three examples of a complete retrospective analysis of the scoring functions to quantify the virtual screening accuracy of the methods. Moreover, only two publications reported some experimental evaluation of the proposed hits until preparing this manuscript. All of these findings led us to carry out a retrospective performance validation of three different docking protocols, through the analysis of their pose prediction and screening accuracy. Surprisingly, we found that even though all tested docking protocols have a good pose prediction, their screening accuracy is quite limited as they fail to correctly rank a test set of compounds. These results highlight the importance of conducting an adequate validation of the docking protocols before carrying out virtual screening campaigns, and to experimentally confirm the predictions made by the models before drawing bold conclusions. Finally, successful structure-based drug discovery investigations published during the redaction of this manuscript allow us to propose the inclusion of target flexibility and consensus scoring as alternatives to improve the accuracy of the methods.
Collapse
Affiliation(s)
- Manuel A Llanos
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), 47&115, La Plata (B1900ADU), Buenos Aires, Argentina
| | - Melisa E Gantner
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), 47&115, La Plata (B1900ADU), Buenos Aires, Argentina
| | - Santiago Rodriguez
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), 47&115, La Plata (B1900ADU), Buenos Aires, Argentina
| | - Lucas N Alberca
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), 47&115, La Plata (B1900ADU), Buenos Aires, Argentina
| | - Carolina L Bellera
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), 47&115, La Plata (B1900ADU), Buenos Aires, Argentina
| | - Alan Talevi
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), 47&115, La Plata (B1900ADU), Buenos Aires, Argentina
| | - Luciana Gavernet
- Laboratory of Bioactive Research and Development (LIDeB), Department of Biological Sciences, Faculty of Exact Sciences, National University of La Plata (UNLP), 47&115, La Plata (B1900ADU), Buenos Aires, Argentina
| |
Collapse
|
11
|
Gossen J, Albani S, Hanke A, Joseph BP, Bergh C, Kuzikov M, Costanzi E, Manelfi C, Storici P, Gribbon P, Beccari AR, Talarico C, Spyrakis F, Lindahl E, Zaliani A, Carloni P, Wade RC, Musiani F, Kokh DB, Rossetti G. A Blueprint for High Affinity SARS-CoV-2 Mpro Inhibitors from Activity-Based Compound Library Screening Guided by Analysis of Protein Dynamics. ACS Pharmacol Transl Sci 2021; 4:1079-1095. [PMID: 34136757 PMCID: PMC8009102 DOI: 10.1021/acsptsci.0c00215] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Indexed: 12/27/2022]
Abstract
The SARS-CoV-2 coronavirus outbreak continues to spread at a rapid rate worldwide. The main protease (Mpro) is an attractive target for anti-COVID-19 agents. Unexpected difficulties have been encountered in the design of specific inhibitors. Here, by analyzing an ensemble of ∼30 000 SARS-CoV-2 Mpro conformations from crystallographic studies and molecular simulations, we show that small structural variations in the binding site dramatically impact ligand binding properties. Hence, traditional druggability indices fail to adequately discriminate between highly and poorly druggable conformations of the binding site. By performing ∼200 virtual screenings of compound libraries on selected protein structures, we redefine the protein's druggability as the consensus chemical space arising from the multiple conformations of the binding site formed upon ligand binding. This procedure revealed a unique SARS-CoV-2 Mpro blueprint that led to a definition of a specific structure-based pharmacophore. The latter explains the poor transferability of potent SARS-CoV Mpro inhibitors to SARS-CoV-2 Mpro, despite the identical sequences of the active sites. Importantly, application of the pharmacophore predicted novel high affinity inhibitors of SARS-CoV-2 Mpro, that were validated by in vitro assays performed here and by a newly solved X-ray crystal structure. These results provide a strong basis for effective rational drug design campaigns against SARS-CoV-2 Mpro and a new computational approach to screen protein targets with malleable binding sites.
Collapse
Affiliation(s)
- Jonas Gossen
- Institute
for Neuroscience and Medicine (INM-9), Forschungszentrum
Jülich, Jülich, 52425, Germany
- Institute
for Advanced Simulations (IAS-5) “Computational biomedicine”, Forschungszentrum Jülich, Jülich, 52425, Germany
- Faculty of
Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, 52062, Germany
| | - Simone Albani
- Institute
for Neuroscience and Medicine (INM-9), Forschungszentrum
Jülich, Jülich, 52425, Germany
- Institute
for Advanced Simulations (IAS-5) “Computational biomedicine”, Forschungszentrum Jülich, Jülich, 52425, Germany
- Faculty of
Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, 52062, Germany
| | - Anton Hanke
- Molecular
and Cellular Modeling Group, Heidelberg
Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, Heidelberg, 69118, Germany
- Institute
of Pharmacy and Molecular Biotechnology (IPMB), Heidelberg University, Im Neuenheimer Feld 364, Heidelberg, 69120, Germany
| | - Benjamin P. Joseph
- Institute
for Neuroscience and Medicine (INM-9), Forschungszentrum
Jülich, Jülich, 52425, Germany
- Institute
for Advanced Simulations (IAS-5) “Computational biomedicine”, Forschungszentrum Jülich, Jülich, 52425, Germany
- Faculty of
Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, 52062, Germany
| | - Cathrine Bergh
- Science for
Life Laboratory & Swedish e-Science Research Center, Department
of Applied Physics, KTH Royal Institute
of Technology, Stockholm, 11428, Sweden
| | - Maria Kuzikov
- Department
of Screening Port, Fraunhofer Institute
for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, Hamburg, 22525, Germany
| | - Elisa Costanzi
- Elettra-Sincrotrone
Trieste S.C.p.A., SS 14-km 163,5 in AREA Science Park, Basovizza,
Trieste, 34149, Italy
| | - Candida Manelfi
- Dompé
Farmaceutici SpA, Via Campo di Pile, L’Aquila, 67100, Italy
| | - Paola Storici
- Elettra-Sincrotrone
Trieste S.C.p.A., SS 14-km 163,5 in AREA Science Park, Basovizza,
Trieste, 34149, Italy
| | - Philip Gribbon
- Department
of Screening Port, Fraunhofer Institute
for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, Hamburg, 22525, Germany
| | | | - Carmine Talarico
- Dompé
Farmaceutici SpA, Via Campo di Pile, L’Aquila, 67100, Italy
| | - Francesca Spyrakis
- Department
of Drug Science and Technology, University
of Turin, via Giuria
9, Turin, 10125, Italy
| | - Erik Lindahl
- Science for
Life Laboratory & Swedish e-Science Research Center, Department
of Applied Physics, KTH Royal Institute
of Technology, Stockholm, 11428, Sweden
- Science
for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, SE-106 91, Sweden
| | - Andrea Zaliani
- Department
of Screening Port, Fraunhofer Institute
for Translational Medicine and Pharmacology ITMP, Schnackenburgallee 114, Hamburg, 22525, Germany
| | - Paolo Carloni
- Institute
for Neuroscience and Medicine (INM-9), Forschungszentrum
Jülich, Jülich, 52425, Germany
- Institute
for Molecular Neuroscience and Neuroimaging (INM-11), Forschungszentrum Jülich, Jülich, 52425, Germany
- Institute
for Advanced Simulations (IAS-5) “Computational biomedicine”, Forschungszentrum Jülich, Jülich, 52425, Germany
- Faculty of
Mathematics, Computer Science and Natural Sciences, RWTH Aachen, Aachen, 52062, Germany
| | - Rebecca C. Wade
- Molecular
and Cellular Modeling Group, Heidelberg
Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, Heidelberg, 69118, Germany
- Zentrum
für Molekulare Biologie der University Heidelberg, DKFZ-ZMBH
Alliance, INF 282, Heidelberg, 69120, Germany
- Interdisciplinary
Center for Scientific Computing (IWR), Heidelberg
University, INF 368, Heidelberg, 69120, Germany
| | - Francesco Musiani
- Laboratory
of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Daria B. Kokh
- Molecular
and Cellular Modeling Group, Heidelberg
Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, Heidelberg, 69118, Germany
| | - Giulia Rossetti
- Institute
for Neuroscience and Medicine (INM-9), Forschungszentrum
Jülich, Jülich, 52425, Germany
- Institute
for Advanced Simulations (IAS-5) “Computational biomedicine”, Forschungszentrum Jülich, Jülich, 52425, Germany
- Jülich
Supercomputing Center (JSC), Forschungszentrum
Jülich, Jülich, 52425, Germany
- Department
of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, RWTH Aachen University, Aachen, 44517, Germany
| |
Collapse
|
12
|
Abstract
The COVID-19 pandemic has generated an unprecedented response within the scientific community. Extraordinary efforts have been undertaken to identify potential new therapeutics to treat SARS-CoV-2 infection spanning traditional medicinal chemistry, repurposing, and computational approaches. The breadth of the effort and rapid progression of many small molecules to clinical testing provide an opportunity to determine what chemical and testing approaches have been the most efficient in identifying potential treatments and how this may inform preparation for future pandemics.
Collapse
Affiliation(s)
- Mark N. Namchuk
- Department of Biological Chemistry and Molecular
Pharmacology, Harvard Medical School, 25
Shattuck Street, Boston, Massachusetts 02115, United
States
| |
Collapse
|
13
|
Anju A, Chaturvedi S, Chaudhary V, Pant P, Hussain F, Mishra AK. Virtual screening of quinoline derived library for SARS-COV-2 targeting viral entry and replication. J Biomol Struct Dyn 2021; 40:8464-8493. [PMID: 34032180 PMCID: PMC8171009 DOI: 10.1080/07391102.2021.1913228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The COVID-19 pandemic infection has claimed many lives and added to the social, economic, and psychological distress. The contagious disease has quickly spread to almost 218 countries and territories following the regional outbreak in China. As the number of infected populations increases exponentially, there is a pressing demand for anti-COVID drugs and vaccines. Virtual screening provides possible leads while extensively cutting down the time and resources required for ab-initio drug design. We report structure-based virtual screening of a hundred plus library of quinoline drugs with established antiviral, antimalarial, antibiotic or kinase inhibitor activity. In this study, targets having a role in viral entry, viral assembly, and viral replication have been selected. The targets include: 1) RBD of receptor-binding domain spike protein S 2) Mpro Chymotrypsin main protease 3) Ppro Papain protease 4) RNA binding domain of Nucleocapsid Protein, and 5) RNA Dependent RNA polymerase from SARS-COV-2. An in-depth analysis of the interactions and G-score compared to the controls like hydroxyquinoline and remdesivir has been presented. The salient results are (1) higher scoring of antivirals as potential drugs (2) potential of afatinib by scoring as better inhibitor, and (3) biological explanation of the potency of afatinib. Further MD simulations and MM-PBSA calculations showed that afatinib works best to interfere with the the activity of RNA dependent RNA polymerase of SARS-COV-2, thereby inhibiting replication process of single stranded RNA virus. Communicated by Ramaswamy H. Sarma
Collapse
Affiliation(s)
- Anju Anju
- Department of Chemistry, University of Delhi, Delhi, India.,Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization Brig, Delhi, India
| | - Shubhra Chaturvedi
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization Brig, Delhi, India
| | - Vishakha Chaudhary
- Department of Chemistry, University of Delhi, Delhi, India.,Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization Brig, Delhi, India
| | - Pradeep Pant
- Department of Chemistry, Indian Institute of Technology, New Delhi, India
| | | | - Anil Kumar Mishra
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization Brig, Delhi, India
| |
Collapse
|
14
|
Rajput A, Thakur A, Mukhopadhyay A, Kamboj S, Rastogi A, Gautam S, Jassal H, Kumar M. Prediction of repurposed drugs for Coronaviruses using artificial intelligence and machine learning. Comput Struct Biotechnol J 2021; 19:3133-3148. [PMID: 34055238 PMCID: PMC8141697 DOI: 10.1016/j.csbj.2021.05.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
The world is facing the COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Likewise, other viruses of the Coronaviridae family were responsible for causing epidemics earlier. To tackle these viruses, there is a lack of approved antiviral drugs. Therefore, we have developed robust computational methods to predict the repurposed drugs using machine learning techniques namely Support Vector Machine, Random Forest, k-Nearest Neighbour, Artificial Neural Network, and Deep Learning. We used the experimentally validated drugs/chemicals with anticorona activity (IC50/EC50) from 'DrugRepV' repository. The unique entries of SARS-CoV-2 (142), SARS (221), MERS (123), and overall Coronaviruses (414) were subdivided into the training/testing and independent validation datasets, followed by the extraction of chemical/structural descriptors and fingerprints (17968). The highly relevant features were filtered using the recursive feature selection algorithm. The selected chemical descriptors were used to develop prediction models with Pearson's correlation coefficients ranging from 0.60 to 0.90 on training/testing. The robustness of the predictive models was further ensured using external independent validation datasets, decoy datasets, applicability domain, and chemical analyses. The developed models were used to predict promising repurposed drug candidates against coronaviruses after scanning the DrugBank. Top predicted molecules for SARS-CoV-2 were further validated by molecular docking against the spike protein complex with ACE receptor. We found potential repurposed drugs namely Verteporfin, Alatrofloxacin, Metergoline, Rescinnamine, Leuprolide, and Telotristat ethyl with high binding affinity. These 'anticorona' computational models would assist in antiviral drug discovery against SARS-CoV-2 and other Coronaviruses.
Collapse
Affiliation(s)
- Akanksha Rajput
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
| | - Anamika Thakur
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Adhip Mukhopadhyay
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sakshi Kamboj
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amber Rastogi
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sakshi Gautam
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Harvinder Jassal
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
| | - Manoj Kumar
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
15
|
Citarella A, Scala A, Piperno A, Micale N. SARS-CoV-2 M pro: A Potential Target for Peptidomimetics and Small-Molecule Inhibitors. Biomolecules 2021; 11:607. [PMID: 33921886 PMCID: PMC8073203 DOI: 10.3390/biom11040607] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
The uncontrolled spread of the COVID-19 pandemic caused by the new coronavirus SARS-CoV-2 during 2020-2021 is one of the most devastating events in the history, with remarkable impacts on the health, economic systems, and habits of the entire world population. While some effective vaccines are nowadays approved and extensively administered, the long-term efficacy and safety of this line of intervention is constantly under debate as coronaviruses rapidly mutate and several SARS-CoV-2 variants have been already identified worldwide. Then, the WHO's main recommendations to prevent severe clinical complications by COVID-19 are still essentially based on social distancing and limitation of human interactions, therefore the identification of new target-based drugs became a priority. Several strategies have been proposed to counteract such viral infection, including the repurposing of FDA already approved for the treatment of HIV, HCV, and EBOLA, inter alia. Among the evaluated compounds, inhibitors of the main protease of the coronavirus (Mpro) are becoming more and more promising candidates. Mpro holds a pivotal role during the onset of the infection and its function is intimately related with the beginning of viral replication. The interruption of its catalytic activity could represent a relevant strategy for the development of anti-coronavirus drugs. SARS-CoV-2 Mpro is a peculiar cysteine protease of the coronavirus family, responsible for the replication and infectivity of the parasite. This review offers a detailed analysis of the repurposed drugs and the newly synthesized molecules developed to date for the treatment of COVID-19 which share the common feature of targeting SARS-CoV-2 Mpro, as well as a brief overview of the main enzymatic and cell-based assays to efficaciously screen such compounds.
Collapse
Affiliation(s)
| | | | | | - Nicola Micale
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, 98166 Messina, Italy; (A.C.); (A.S.); (A.P.)
| |
Collapse
|
16
|
Fischer A, Sellner M, Mitusińska K, Bzówka M, Lill MA, Góra A, Smieško M. Computational Selectivity Assessment of Protease Inhibitors against SARS-CoV-2. Int J Mol Sci 2021; 22:2065. [PMID: 33669738 PMCID: PMC7922391 DOI: 10.3390/ijms22042065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 12/27/2022] Open
Abstract
The pandemic of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a serious global health threat. Since no specific therapeutics are available, researchers around the world screened compounds to inhibit various molecular targets of SARS-CoV-2 including its main protease (Mpro) essential for viral replication. Due to the high urgency of these discovery efforts, off-target binding, which is one of the major reasons for drug-induced toxicity and safety-related drug attrition, was neglected. Here, we used molecular docking, toxicity profiling, and multiple molecular dynamics (MD) protocols to assess the selectivity of 33 reported non-covalent inhibitors of SARS-CoV-2 Mpro against eight proteases and 16 anti-targets. The panel of proteases included SARS-CoV Mpro, cathepsin G, caspase-3, ubiquitin carboxy-terminal hydrolase L1 (UCHL1), thrombin, factor Xa, chymase, and prostasin. Several of the assessed compounds presented considerable off-target binding towards the panel of proteases, as well as the selected anti-targets. Our results further suggest a high risk of off-target binding to chymase and cathepsin G. Thus, in future discovery projects, experimental selectivity assessment should be directed toward these proteases. A systematic selectivity assessment of SARS-CoV-2 Mpro inhibitors, as we report it, was not previously conducted.
Collapse
Affiliation(s)
- André Fischer
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| | - Manuel Sellner
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| | - Karolina Mitusińska
- Tunneling Group, Biotechnology Centre, ul. Krzywoustego 8, Silesian University of Technology, 44-100 Gliwice, Poland; (K.M.); (M.B.)
| | - Maria Bzówka
- Tunneling Group, Biotechnology Centre, ul. Krzywoustego 8, Silesian University of Technology, 44-100 Gliwice, Poland; (K.M.); (M.B.)
| | - Markus A. Lill
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| | - Artur Góra
- Tunneling Group, Biotechnology Centre, ul. Krzywoustego 8, Silesian University of Technology, 44-100 Gliwice, Poland; (K.M.); (M.B.)
| | - Martin Smieško
- Computational Pharmacy, Departement of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (A.F.); (M.S.)
| |
Collapse
|
17
|
Chen CZ, Shinn P, Itkin Z, Eastman RT, Bostwick R, Rasmussen L, Huang R, Shen M, Hu X, Wilson KM, Brooks BM, Guo H, Zhao T, Klump-Thomas C, Simeonov A, Michael SG, Lo DC, Hall MD, Zheng W. Drug Repurposing Screen for Compounds Inhibiting the Cytopathic Effect of SARS-CoV-2. Front Pharmacol 2021; 11:592737. [PMID: 33708112 PMCID: PMC7942396 DOI: 10.3389/fphar.2020.592737] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/04/2020] [Indexed: 11/23/2022] Open
Abstract
Drug repurposing is a rapid approach to identify therapeutics for the treatment of emerging infectious diseases such as COVID-19. To address the urgent need for treatment options, we carried out a quantitative high-throughput screen using a SARS-CoV-2 cytopathic assay with a compound collection of 8,810 approved and investigational drugs, mechanism-based bioactive compounds, and natural products. Three hundred and nineteen compounds with anti-SARS-CoV-2 activities were identified and confirmed, including 91 approved drugs and 49 investigational drugs. The anti-SARS-CoV-2 activities of 230 of these confirmed compounds, of which 38 are approved drugs, have not been previously reported. Chlorprothixene, methotrimeprazine, and piperacetazine were the three most potent FDA-approved drugs with anti-SARS-CoV-2 activities. These three compounds have not been previously reported to have anti-SARS-CoV-2 activities, although their antiviral activities against SARS-CoV and Ebola virus have been reported. These results demonstrate that this comprehensive data set is a useful resource for drug repurposing efforts, including design of new drug combinations for clinical trials for SARS-CoV-2.
Collapse
Affiliation(s)
- Catherine Z. Chen
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Paul Shinn
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Zina Itkin
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Richard T. Eastman
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | | | | | - Ruili Huang
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Min Shen
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Xin Hu
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Kelli M. Wilson
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Brianna M. Brooks
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Hui Guo
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Tongan Zhao
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Carleen Klump-Thomas
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Samuel G. Michael
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Donald C. Lo
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Matthew D. Hall
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| | - Wei Zheng
- National Center for Advancing Translational Sciences, Rockville, MD, United States
| |
Collapse
|
18
|
The role of chemical biology in the fight against SARS-CoV-2. Biochem J 2021; 478:157-177. [PMID: 33439990 DOI: 10.1042/bcj20200514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 01/18/2023]
Abstract
Since late 2019, biomedical labs all over the world have been struggling to cope with the 'new normal' and to find ways in which they can contribute to the fight against COVID-19. In this unique situation where a biomedical issue dominates people's lives and the news cycle, chemical biology has a great deal to contribute. This review will describe the importance of science at the chemistry/biology interface to both understand and combat the SARS-CoV-2 pandemic.
Collapse
|
19
|
Ghahremanpour MM, Tirado-Rives J, Deshmukh M, Ippolito JA, Zhang CH, Cabeza de Vaca I, Liosi ME, Anderson KS, Jorgensen WL. Identification of 14 Known Drugs as Inhibitors of the Main Protease of SARS-CoV-2. ACS Med Chem Lett 2020; 11:2526-2533. [PMID: 33324471 PMCID: PMC7605328 DOI: 10.1021/acsmedchemlett.0c00521] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
![]()
A consensus virtual screening protocol has been applied to ca.
2000 approved drugs to seek inhibitors of the main protease (Mpro) of SARS-CoV-2, the virus responsible for COVID-19. 42
drugs emerged as top candidates, and after visual analyses of the
predicted structures of their complexes with Mpro, 17 were
chosen for evaluation in a kinetic assay for Mpro inhibition.
Remarkably 14 of the compounds at 100-μM concentration were
found to reduce the enzymatic activity and 5 provided IC50 values below 40 μM: manidipine (4.8 μM), boceprevir
(5.4 μM), lercanidipine (16.2 μM), bedaquiline (18.7 μM),
and efonidipine (38.5 μM). Structural analyses reveal a common
cloverleaf pattern for the binding of the active compounds to the
P1, P1′, and P2 pockets of Mpro. Further study of
the most active compounds in the context of COVID-19 therapy is warranted,
while all of the active compounds may provide a foundation for lead
optimization to deliver valuable chemotherapeutics to combat the pandemic.
Collapse
Affiliation(s)
| | - Julian Tirado-Rives
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States,
| | - Maya Deshmukh
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8066, United States and
| | - Joseph A. Ippolito
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States,
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8066, United States and
| | - Chun-Hui Zhang
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States,
| | - Israel Cabeza de Vaca
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States,
| | - Maria-Elena Liosi
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States,
| | - Karen S. Anderson
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8066, United States and
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520-8066, United States
| | - William L. Jorgensen
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States,
| |
Collapse
|
20
|
Bolcato G, Bissaro M, Pavan M, Sturlese M, Moro S. Targeting the coronavirus SARS-CoV-2: computational insights into the mechanism of action of the protease inhibitors lopinavir, ritonavir and nelfinavir. Sci Rep 2020; 10:20927. [PMID: 33262359 PMCID: PMC7708625 DOI: 10.1038/s41598-020-77700-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 11/05/2020] [Indexed: 01/30/2023] Open
Abstract
Coronavirus SARS-CoV-2 is a recently discovered single-stranded RNA betacoronavirus, responsible for a severe respiratory disease known as coronavirus disease 2019, which is rapidly spreading. Chinese health authorities, as a response to the lack of an effective therapeutic strategy, started to investigate the use of lopinavir and ritonavir, previously optimized for the treatment and prevention of HIV/AIDS viral infection. Despite the clinical use of these two drugs, no information regarding their possible mechanism of action at the molecular level is still known for SARS-CoV-2. Very recently, the crystallographic structure of the SARS-CoV-2 main protease (Mpro), also known as C30 Endopeptidase, was published. Starting from this essential structural information, in the present work we have exploited supervised molecular dynamics, an emerging computational technique that allows investigating at an atomic level the recognition process of a ligand from its unbound to the final bound state. In this research, we provided molecular insight on the whole recognition pathway of Lopinavir, Ritonavir, and Nelfinavir, three potential C30 Endopeptidase inhibitors, with the last one taken into consideration due to the promising in-vitro activity shown against the structurally related SARS-CoV protease.
Collapse
Affiliation(s)
- Giovanni Bolcato
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Maicol Bissaro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Matteo Pavan
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131, Padova, Italy.
| |
Collapse
|
21
|
Ghahremanpour MM, Tirado-Rives J, Deshmukh M, Ippolito JA, Zhang CH, de Vaca IC, Liosi ME, Anderson KS, Jorgensen WL. Identification of 14 Known Drugs as Inhibitors of the Main Protease of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32869018 DOI: 10.1101/2020.08.28.271957] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A consensus virtual screening protocol has been applied to ca. 2000 approved drugs to seek inhibitors of the main protease (M pro ) of SARS-CoV-2, the virus responsible for COVID-19. 42 drugs emerged as top candidates, and after visual analyses of the predicted structures of their complexes with M pro , 17 were chosen for evaluation in a kinetic assay for M pro inhibition. Remarkably 14 of the compounds at 100-μM concentration were found to reduce the enzymatic activity and 5 provided IC 50 values below 40 μM: manidipine (4.8 μM), boceprevir (5.4 μM), lercanidipine (16.2 μM), bedaquiline (18.7 μM), and efonidipine (38.5 μM). Structural analyses reveal a common cloverleaf pattern for the binding of the active compounds to the P1, P1', and P2 pockets of M pro . Further study of the most active compounds in the context of COVID-19 therapy is warranted, while all of the active compounds may provide a foundation for lead optimization to deliver valuable chemotherapeutics to combat the pandemic.
Collapse
|
22
|
Sencanski M, Perovic V, Pajovic SB, Adzic M, Paessler S, Glisic S. Drug Repurposing for Candidate SARS-CoV-2 Main Protease Inhibitors by a Novel In Silico Method. Molecules 2020; 25:E3830. [PMID: 32842509 PMCID: PMC7503980 DOI: 10.3390/molecules25173830] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/05/2020] [Accepted: 08/21/2020] [Indexed: 01/01/2023] Open
Abstract
The SARS-CoV-2 outbreak caused an unprecedented global public health threat, having a high transmission rate with currently no drugs or vaccines approved. An alternative powerful additional approach to counteract COVID-19 is in silico drug repurposing. The SARS-CoV-2 main protease is essential for viral replication and an attractive drug target. In this study, we used the virtual screening protocol with both long-range and short-range interactions to select candidate SARS-CoV-2 main protease inhibitors. First, the Informational spectrum method applied for small molecules was used for searching the Drugbank database and further followed by molecular docking. After in silico screening of drug space, we identified 57 drugs as potential SARS-CoV-2 main protease inhibitors that we propose for further experimental testing.
Collapse
Affiliation(s)
- Milan Sencanski
- Laboratory of Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences Vinca, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia; (M.S.); (V.P.)
| | - Vladimir Perovic
- Laboratory of Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences Vinca, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia; (M.S.); (V.P.)
| | - Snezana B. Pajovic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia; (S.B.P.); (M.A.)
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia; (S.B.P.); (M.A.)
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA;
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Sanja Glisic
- Laboratory of Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences Vinca, National Institute of the Republic of Serbia, University of Belgrade, 11001 Belgrade, Serbia; (M.S.); (V.P.)
| |
Collapse
|
23
|
Palma G, Pasqua T, Silvestri G, Rocca C, Gualtieri P, Barbieri A, De Bartolo A, De Lorenzo A, Angelone T, Avolio E, Botti G. PI3Kδ Inhibition as a Potential Therapeutic Target in COVID-19. Front Immunol 2020; 11:2094. [PMID: 32973818 PMCID: PMC7472874 DOI: 10.3389/fimmu.2020.02094] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/03/2020] [Indexed: 01/08/2023] Open
Abstract
The spread of the novel human respiratory coronavirus (SARS-CoV-2) is a global public health emergency. There is no known successful treatment as of this time, and there is a need for medical options to mitigate this current epidemic. SARS-CoV-2 uses the angiotensin-converting enzyme 2 (ACE2) receptor and is primarily trophic for the lower and upper respiratory tract. A number of current studies on COVID-19 have demonstrated the substantial increase in pro-inflammatory factors in the lungs during infection. The virus is also documented in the central nervous system and, particularly in the brainstem, which plays a key role in respiratory and cardiovascular function. Currently, there are few antiviral approaches, and several alternative drugs are under investigation. Two of these are Idelalisib and Ebastine, already proposed as preventive strategies in airways and allergic diseases. The interesting and evolving potential of phosphoinositide 3-kinase δ (PI3Kδ) inhibitors, together with Ebastine, lies in their ability to suppress the release of pro-inflammatory cytokines, such as IL-1β, IL-8, IL-6, and TNF-α, by T cells. This may represent an optional therapeutic choice for COVID-19 to reduce inflammatory reactions and mortality, enabling patients to recover faster. This concise communication aims to provide new potential therapeutic targets capable of mitigating and alleviating SARS-CoV-2 pandemic infection.
Collapse
Affiliation(s)
- Giuseppe Palma
- SSD Sperimentazione Animale, Istituto Nazionale Tumori Fondazione G. Pascale – IRCSS, Naples, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Giovannino Silvestri
- Institute of Human Virology, Division of Infectious Agents and Cancer, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Paola Gualtieri
- School of Specialization in Food Science, University of Rome “Tor Vergata”, Rome, Italy
| | - Antonio Barbieri
- SSD Sperimentazione Animale, Istituto Nazionale Tumori Fondazione G. Pascale – IRCSS, Naples, Italy
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - Antonino De Lorenzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Patho-Physiology, Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
- National Institute for Cardiovascular Research (INRC), Bologna, Italy
| | - Ennio Avolio
- School of Specialization in Food Science, University of Rome “Tor Vergata”, Rome, Italy
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Gerardo Botti
- Scientific Director, Istituto Nazionale Tumori Fondazione G. Pascale – IRCSS, Naples, Italy
| |
Collapse
|
24
|
Li Q, Kang C. Progress in Developing Inhibitors of SARS-CoV-2 3C-Like Protease. Microorganisms 2020; 8:E1250. [PMID: 32824639 PMCID: PMC7463875 DOI: 10.3390/microorganisms8081250] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The viral outbreak started in late 2019 and rapidly became a serious health threat to the global population. COVID-19 was declared a pandemic by the World Health Organization in March 2020. Several therapeutic options have been adopted to prevent the spread of the virus. Although vaccines have been developed, antivirals are still needed to combat the infection of this virus. SARS-CoV-2 is an enveloped virus, and its genome encodes polyproteins that can be processed into structural and nonstructural proteins. Maturation of viral proteins requires cleavages by proteases. Therefore, the main protease (3 chymotrypsin-like protease (3CLpro) or Mpro) encoded by the viral genome is an attractive drug target because it plays an important role in cleaving viral polyproteins into functional proteins. Inhibiting this enzyme is an efficient strategy to block viral replication. Structural studies provide valuable insight into the function of this protease and structural basis for rational inhibitor design. In this review, we describe structural studies on the main protease of SARS-CoV-2. The strategies applied in developing inhibitors of the main protease of SARS-CoV-2 and currently available protein inhibitors are summarized. Due to the availability of high-resolution structures, structure-guided drug design will play an important role in developing antivirals. The availability of high-resolution structures, potent peptidic inhibitors, and diverse compound scaffolds indicate the feasibility of developing potent protease inhibitors as antivirals for COVID-19.
Collapse
Affiliation(s)
- Qingxin Li
- Guangdong Provincial Engineering Laboratory of Biomass High Value Utilization, Institute of Bioengineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - CongBao Kang
- Experimental Drug Development Centre (EDDC), Agency for Science, Technology and Research (A*STAR), 10 Biopolis Road, Chromos, #05-01, Singapore 138670, Singapore
| |
Collapse
|
25
|
Chen CZ, Shinn P, Itkin Z, Eastman RT, Bostwick R, Rasmussen L, Huang R, Shen M, Hu X, Wilson KM, Brooks B, Guo H, Zhao T, Klump-Thomas C, Simeonov A, Michael SG, Lo DC, Hall MD, Zheng W. Drug Repurposing Screen for Compounds Inhibiting the Cytopathic Effect of SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.08.18.255877. [PMID: 32839771 PMCID: PMC7444282 DOI: 10.1101/2020.08.18.255877] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug repurposing is a rapid approach to identifying therapeutics for the treatment of emerging infectious diseases such as COVID-19. To address the urgent need for treatment options, we carried out a quantitative high-throughput screen using a SARS-CoV-2 cytopathic assay with a compound collection of 8,810 approved and investigational drugs, mechanism-based bioactive compounds, and natural products. Three hundred and nineteen compounds with anti-SARS-CoV-2 activities were identified and confirmed, including 91 approved drug and 49 investigational drugs. Among these confirmed compounds, the anti-SARS-CoV-2 activities of 230 compounds, including 38 approved drugs, have not been previously reported. Chlorprothixene, methotrimeprazine, and piperacetazine were the three most potent FDA approved drugs with anti-SARS-CoV-2 activities. These three compounds have not been previously reported to have anti-SARS-CoV-2 activities, although their antiviral activities against SARS-CoV and Ebola virus have been reported. These results demonstrate that this comprehensive data set of drug repurposing screen for SARS-CoV-2 is useful for drug repurposing efforts including design of new drug combinations for clinical trials.
Collapse
Affiliation(s)
- Catherine Z. Chen
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Paul Shinn
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Zina Itkin
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Richard T. Eastman
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Robert Bostwick
- Southern Research, 2000 Ninth Avenue South, Birmingham, Alabama, 35205
| | - Lynn Rasmussen
- Southern Research, 2000 Ninth Avenue South, Birmingham, Alabama, 35205
| | - Ruili Huang
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Min Shen
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Xin Hu
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Kelli M. Wilson
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Brianna Brooks
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Hui Guo
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Tongan Zhao
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Carleen Klump-Thomas
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Samuel G. Michael
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Donald C. Lo
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Matthew D. Hall
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| | - Wei Zheng
- National Center for Advancing Translational Sciences, 9800 Medical Center Drive, Rockville, MD, 20850
| |
Collapse
|