Discovery and Mechanism of SARS-CoV-2 Main Protease InhibitorsClick to copy article linkArticle link copied!
- Sarah HuffSarah HuffDivision of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Sarah Huff
- Indrasena Reddy KummethaIndrasena Reddy KummethaDivision of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Indrasena Reddy Kummetha
- Shashi Kant TiwariShashi Kant TiwariDivision of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Shashi Kant Tiwari
- Matthew B. HuanteMatthew B. HuanteDepartment of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United StatesMore by Matthew B. Huante
- Alex E. ClarkAlex E. ClarkDivision of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Alex E. Clark
- Shaobo WangShaobo WangDivision of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Shaobo Wang
- William BrayWilliam BrayDivision of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by William Bray
- Davey SmithDavey SmithDivision of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Davey Smith
- Aaron F. CarlinAaron F. CarlinDivision of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Aaron F. Carlin
- Mark EndsleyMark EndsleyDepartment of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United StatesMore by Mark Endsley
- Tariq M. Rana*Tariq M. Rana*Email: [email protected]Division of Genetics, Department of Pediatrics, Center for Drug Discovery Innovation, Program in Immunology, Institute for Genomic Medicine, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesDivision of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, 9500 Gilman Drive MC 0762, La Jolla, California 92093, United StatesMore by Tariq M. Rana
Abstract
The emergence of a new coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents an urgent public health crisis. Without available targeted therapies, treatment options remain limited for COVID-19 patients. Using medicinal chemistry and rational drug design strategies, we identify a 2-phenyl-1,2-benzoselenazol-3-one class of compounds targeting the SARS-CoV-2 main protease (Mpro). FRET-based screening against recombinant SARS-CoV-2 Mpro identified six compounds that inhibit proteolysis with nanomolar IC50 values. Preincubation dilution experiments and molecular docking determined that the inhibition of SARS-CoV-2 Mpro can occur by either covalent or noncovalent mechanisms, and lead E04 was determined to inhibit Mpro competitively. Lead E24 inhibited viral replication with a nanomolar EC50 value (844 nM) in SARS-CoV-2-infected Vero E6 cells and was further confirmed to impair SARS-CoV-2 replication in human lung epithelial cells and human-induced pluripotent stem cell-derived 3D lung organoids. Altogether, these studies provide a structural framework and mechanism of Mpro inhibition that should facilitate the design of future COVID-19 treatments.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
SPECIAL ISSUE
This article is part of the
Introduction
Results
Identification of the 2-Phenyl-1,2-benzoselenazol-3-one Class of SARS-CoV-2 Mpro Inhibitors
The 2-Phenyl-1,2-benzoselenazol-3-one Class of Compounds Are Potent Inhibitors of Recombinant Mpro
2-Phenyl-1,2-benzoselenazol-3-one Inhibitors Can Inhibit Mpro Covalently or Noncovalently
2-Phenyl-1,2-benzoselenazol-3-one Inhibitor E04 and Ebselen Are Competitive Inhibitors of Mpro
2-Phenyl-1,2-benzoselenazol-3-one Analogues Are Potent Antiviral Agents In Vitro
compound | antiviral EC50 (μM) | logD pH 7.4 | compound | antiviral EC50 (μM) | logD pH 7.4 |
---|---|---|---|---|---|
ebselen | >20 | 0.92 | remdesivir | 1.8 ± 1.2 | ND |
E04 | 11.2 ± 1.3 | 3.19 | E20 | 18.2 ± 3.6 | 1.67 |
E07 | 26.5 ± 1.2 | 0.85 | E21 | 5.2 ± 1.8 | 1.46 |
E18 | 6.5 ± 2.0 | 2.87 | E24 | 0.8 ± 0.3 | 3.18 |
E19 | 17.4 ± 3.5 | 1.53 | E25 | 2.0 ± 1.1 | 1.45 |
ND, not determined.
2-Phenyl-1,2-benzoselenazol-3-one Compounds Show Improved LogD Values Relative to Ebselen
2-Phenyl-1,2-benzoselenazol-3-one Analogues Show Antiviral Properties in Human Lung Cells
compound | EC50 (μM) | compound | EC50 (μM) |
---|---|---|---|
ebselen | 5.0 ± 4.0 | remdesivir | 0.8 ± 0.6 |
E04 | 2.8 ± 1.3 | E21 | 15.7 ± 4.3 |
E18 | 8.8 ± 5.4 | E24 | 1.3 ± 0.8 |
Compound E24 Reduces SARS-CoV-2 Viral Infection in Human Lung Organoids
Discussion and Conclusions
Experimental Section
In Silico Modeling of Ebselen Analogues with Schrödinger
SARS-CoV-2 Main Protease Protein Expression and Purification
FRET Protease Activity Assay
Inhibition Mechanism
logD Measurements
Antiviral Assays
Lung Organoid Infection and Treatment with E24
General Procedure for the Synthesis of Compounds E01–30
Compound Characterization
2-Phenylbenzo[d][1,2]selenazol-3(2H)-one (E01)
2-(4-Fluorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E02)
2-(2-(Methylthio)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E03)
2-(4-(Trifluoromethyl)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E04)
2-(3,4-Dimethylphenyl)benzo[d][1,2]selenazol-3(2H)-one (E05)
2-(4-Fluorobenzyl)benzo[d][1,2]selenazol-3(2H)-one (E06)
2-(2,4-Dimethoxybenzyl)benzo[d][1,2]selenazol-3(2H)-one (E07)
2-(2-Fluorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E08)
2-(4-(Trifluoromethyl)benzyl)benzo[d][1,2]selenazol-3(2H)-one (E09)
2-(4-Fluoro-2-(trifluoromethyl)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E10)
2-(2,6-Difluorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E11)
2-(4-Methoxybenzyl)benzo[d][1,2]selenazol-3(2H)-one (E12)
2-(4-Chloro-2-(trifluoromethoxy)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E13)
2-(5-Chloro-2-methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E14)
2-(4-Chlorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E15)
2-(5-Fluoro-2-methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E16)
2-(4-Fluoro-2-methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E17)
2-(4-Ethylphenyl)benzo[d][1,2]selenazol-3(2H)-one (E18)
2-(3-Chlorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E19)
2-(3,5-Dichlorophenyl)benzo[d][1,2]selenazol-3(2H)-one (E20)
2-(4-Bromophenyl)benzo[d][1,2]selenazol-3(2H)-one (E21)
2-(p-Tolyl)benzo[d][1,2]selenazol-3(2H)-one (E22)
2-(o-Tolyl)benzo[d][1,2]selenazol-3(2H)-one (E23)
2-(3-(Trifluoromethyl)phenyl)benzo[d][1,2]selenazol-3(2H)-one (E24)
3-(3-Oxobenzo[d][1,2]selenazol-2(3H)-yl)benzonitrile (E25)
2-(3-Methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E26)
2-(4-Methoxyphenyl)benzo[d][1,2]selenazol-3(2H)-one (E27)
2-Heptylbenzo[d][1,2]selenazol-3(2H)-one (E28)
2-Octylbenzo[d][1,2]selenazol-3(2H)-one (E29)
2-Pentylbenzo[d][1,2]selenazol-3(2H)-one (E30)
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c00566.
Molecular formula strings(CSV)
Expanded synthetic methods and spectra for E01–30, structures and reported activities for known SARS-CoV-2 main protease inhibitors, docking poses, and individual antiviral dose–response curves for select compounds
(PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We thank the Rao Zihe and Yang Haitao laboratories of ShanghaiTech University for providing the coordinates of the SARS-CoV-2 Mpro crystal structure (PDB ID 6UL7). We thank members of the Rana lab for helpful discussions and advice. This work was supported by a T32 fellowship from the National Cancer Institute of the National Institutes of Health (award no. T32CA121938 to S.H.), a Career Award for Medical Scientists from the Burroughs Wellcome Fund (USA), a grant from the National Institutes of Health USA (K08 AI130381) to A.F.C., the John and Mary Tu Foundation (USA), a CIRM award to T.M.R., and in part by grants from the National Institutes of Health (USA) (CA177322, DA039562, DA049524, and AI125103).
ACE2 | angiotensin-converting enzyme 2 |
COVID-19 | coronavirus disease 2019 |
iPSC | induced pluripotent stem cells |
ISG15 | interferon-stimulated gene 15 |
MOI | multiplicity of infection |
Mpro | main protease |
nsp1 | nonstructural protein 1 |
nsp2 | nonstructural protein 2 |
nsp3 | nonstructural protein 3 |
OPLS | optimized potentials for liquid simulations |
PLpro | papain-like protease |
RT-qPCR | real-time quantitative reverse transcription PCR |
RdRp | RNA-dependent RNA polymerase |
SARS-CoV-2 | severe acute respiratory syndrome coronavirus 2 |
SGB | surface generalized born |
(+)ssRNA | positive-sense single-stranded RNA |
TMPRSS2 | transmembrane protease serine 2 |
References
This article references 50 other publications.
- 1Li, F.; Li, W.; Farzan, M.; Harrison, S. C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309 (5742), 1864– 1868, DOI: 10.1126/science.1116480Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpvFCisLw%253D&md5=0a7ac2fb7a76c97979d859c0489c682fStructure of SARS Coronavirus Spike Receptor-Binding Domain Complexed with ReceptorLi, Fang; Li, Wenhui; Farzan, Michael; Harrison, Stephen C.Science (Washington, DC, United States) (2005), 309 (5742), 1864-1868CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The spike protein (S) of SARS coronavirus (SARS-CoV) attaches the virus to its cellular receptor, angiotensin-converting enzyme 2 (ACE2). A defined receptor-binding domain (RBD) on S mediates this interaction. The crystal structure at 2.9 angstrom resoln. of the RBD bound with the peptidase domain of human ACE2 shows that the RBD presents a gently concave surface, which cradles the N-terminal lobe of the peptidase. The at. details at the interface between the two proteins clarify the importance of residue changes that facilitate efficient cross-species infection and human-to-human transmission. The structure of the RBD suggests ways to make truncated disulfide-stabilized RBD variants for use in the design of coronavirus vaccines.
- 2Phan, T. Novel coronavirus: From discovery to clinical diagnostics. Infect., Genet. Evol. 2020, 79, 104211, DOI: 10.1016/j.meegid.2020.104211Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFOit7g%253D&md5=3cb3ef734c4f4f85479d8b55a2daf94cNovel coronavirus: From discovery to clinical diagnosticsPhan, TungInfection, Genetics and Evolution (2020), 79 (), 104211CODEN: IGENCN; ISSN:1567-1348. (Elsevier B.V.)A novel coronavirus designated as 2019-nCoV first appeared in Wuhan, China in late Dec. 2019. Dozens of people died in China, and thousands of people infected as 2019-nCoV continues to spread around the world. We have described the discovery, emergence, genomic characteristics, and clin. diagnostics of 2019-nCoV.
- 3Pillaiyar, T.; Manickam, M.; Namasivayam, V.; Hayashi, Y.; Jung, S. H. An Overview of Severe Acute Respiratory Syndrome-Coronavirus (SARS-CoV) 3CL Protease Inhibitors: Peptidomimetics and Small Molecule Chemotherapy. J. Med. Chem. 2016, 59 (14), 6595– 628, DOI: 10.1021/acs.jmedchem.5b01461Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisVyns7c%253D&md5=6ce37076204280f64a7f362633fcee1aAn Overview of Severe Acute Respiratory Syndrome-Coronavirus (SARS-CoV) 3CL Protease Inhibitors: Peptidomimetics and Small Molecule ChemotherapyPillaiyar, Thanigaimalai; Manickam, Manoj; Namasivayam, Vigneshwaran; Hayashi, Yoshio; Jung, Sang-HunJournal of Medicinal Chemistry (2016), 59 (14), 6595-6628CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Severe acute respiratory syndrome (SARS) is caused by a newly emerged coronavirus that infected more than 8000 individuals and resulted in more than 800 (10-15%) fatalities in 2003. The causative agent of SARS has been identified as a novel human coronavirus (SARS-CoV), and its viral protease, SARS-CoV 3CLpro, has been shown to be essential for replication and has hence been recognized as a potent drug target for SARS infection. Currently, there is no effective treatment for this epidemic despite the intensive research that has been undertaken since 2003 (over 3500 publications). This perspective focuses on the status of various efficacious anti-SARS-CoV 3CLpro chemotherapies discovered during the last 12 years (2003-2015) from all sources, including lab. synthetic methods, natural products, and virtual screening. We describe here mainly peptidomimetic and small mol. inhibitors of SARS-CoV 3CLpro. Attempts have been made to provide a complete description of the structural features and binding modes of these inhibitors under many conditions.
- 4Ziebuhr, J.; Snijder, E. J.; Gorbalenya, A. E. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J. Gen. Virol. 2000, 81 (4), 853– 879, DOI: 10.1099/0022-1317-81-4-853Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXisVektLg%253D&md5=5293084b82548029518d57e401eccc3eVirus-encoded proteinases and proteolytic processing in the nidoviralesZiebuhr, John; Snijder, Eric J.; Gorbalenya, Alexander E.Journal of General Virology (2000), 81 (4), 853-879CODEN: JGVIAY; ISSN:0022-1317. (Society for General Microbiology)A review with 128 refs. The review is organized into five main sections. In the first section nidovirus proteinases are introduced and classified into main and accessory proteinases. In the next two sections a brief overview of two classes of nidovirus proteinases is presented and, in this context, the coronavirus and arterivirus enzymes are compared to each other and to the prototypic proteinases. This is followed by a detailed description of the nidovirus proteinases themselves. The review is concluded by two sections that describe the regulatory role of the proteinases during virus replication and then give an outline of future perspectives concerning nidovirus proteinases.
- 5Báez-Santos, Y. M.; St. John, S. E.; Mesecar, A. D. The SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compounds. Antiviral Res. 2015, 115, 21– 38, DOI: 10.1016/j.antiviral.2014.12.015Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmtVOnsw%253D%253D&md5=fcdde62e05b499546c73f3014ea521adThe SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compoundsBaez-Santos, Yahira M.; St. John, Sarah E.; Mesecar, Andrew D.Antiviral Research (2015), 115 (), 21-38CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)A review. Over 10 years have passed since the deadly human coronavirus that causes severe acute respiratory syndrome (SARS-CoV) emerged from the Guangdong Province of China. Despite the fact that the SARS-CoV pandemic infected over 8500 individuals, claimed over 800 lives and cost billions of dollars in economic loss worldwide, there still are no clin. approved antiviral drugs, vaccines or monoclonal antibody therapies to treat SARS-CoV infections. The recent emergence of the deadly human coronavirus that causes Middle East respiratory syndrome (MERS-CoV) is a sobering reminder that new and deadly coronaviruses can emerge at any time with the potential to become pandemics. Therefore, the continued development of therapeutic and prophylactic countermeasures to potentially deadly coronaviruses is warranted. The coronaviral proteases, papain-like protease (PLpro) and 3C-like protease (3CLpro), are attractive antiviral drug targets because they are essential for coronaviral replication. Although the primary function of PLpro and 3CLpro are to process the viral polyprotein in a coordinated manner, PLpro has the addnl. function of stripping ubiquitin and ISG15 from host-cell proteins to aid coronaviruses in their evasion of the host innate immune responses. Therefore, targeting PLpro with antiviral drugs may have an advantage in not only inhibiting viral replication but also inhibiting the dysregulation of signaling cascades in infected cells that may lead to cell death in surrounding, uninfected cells. This review provides an up-to-date discussion on the SARS-CoV papain-like protease including a brief overview of the SARS-CoV genome and replication followed by a more in-depth discussion on the structure and catalytic mechanism of SARS-CoV PLpro, the multiple cellular functions of SARS-CoV PLpro, the inhibition of SARS-CoV PLpro by small mol. inhibitors, and the prospect of inhibiting papain-like protease from other coronaviruses. This paper forms part of a series of invited articles in Antiviral Research on "From SARS to MERS: 10 years of research on highly pathogenic human coronaviruses.".
- 6Lee, T. W.; Cherney, M. M.; Huitema, C.; Liu, J.; James, K. E.; Powers, J. C.; Eltis, L. D.; James, M. N. Crystal structures of the main peptidase from the SARS coronavirus inhibited by a substrate-like aza-peptide epoxide. J. Mol. Biol. 2005, 353 (5), 1137– 1151, DOI: 10.1016/j.jmb.2005.09.004Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtFKms7nL&md5=ba0e5fbad36bfc3d259c9221d451ef60Crystal Structures of the Main Peptidase from the SARS Coronavirus Inhibited by a Substrate-like Aza-peptide EpoxideLee, Ting-Wai; Cherney, Maia M.; Huitema, Carly; Liu, Jie; James, Karen Ellis; Powers, James C.; Eltis, Lindsay D.; James, Michael N. G.Journal of Molecular Biology (2005), 353 (5), 1137-1151CODEN: JMOBAK; ISSN:0022-2836. (Elsevier B.V.)The main peptidase (Mpro) from the coronavirus (CoV) causing severe acute respiratory syndrome (SARS) is one of the most attractive mol. targets for the development of anti-SARS agents. We report the irreversible inhibition of SARS-CoV Mpro by an aza-peptide epoxide (APE; kinact/Ki=1900(±400) M-1 s-1). The crystal structures of the Mpro:APE complex in the space groups C2 and P212121 revealed the formation of a covalent bond between the catalytic Cys145 Sγ atom of the peptidase and the epoxide C3 atom of the inhibitor, substantiating the mode of action of this class of cysteine-peptidase inhibitors. The aza-peptide component of APE binds in the substrate-binding regions of Mpro in a substrate-like manner, with excellent structural and chem. complementarity. In addn., the crystal structure of unbound Mpro in the space group C2 revealed that the "N-fingers" (N-terminal residues 1 to 7) of both protomers of Mpro are well defined and the substrate-binding regions of both protomers are in the catalytically competent conformation at the crystn. pH of 6.5, contrary to the previously detd. crystal structures of unbound Mpro in the space group P21.
- 7Xu, X.; Liu, Y.; Weiss, S.; Arnold, E.; Sarafianos, S. G.; Ding, J. Molecular model of SARS coronavirus polymerase: implications for biochemical functions and drug design. Nucleic Acids Res. 2003, 31 (24), 7117– 7130, DOI: 10.1093/nar/gkg916Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXps1Siu7Y%253D&md5=1c63b9eb468d4b067d4efb3136e18e66Molecular model of SARS coronavirus polymerase: implications for biochemical functions and drug designXu, Xiang; Liu, Yunqing; Weiss, Susan; Arnold, Eddy; Sarafianos, Stefan G.; Ding, JianpingNucleic Acids Research (2003), 31 (24), 7117-7130CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The causative agent of severe acute respiratory syndrome (SARS) is a previously unidentified coronavirus, SARS-CoV. The RNA-dependent RNA polymerase (RdRp) of SARS-CoV plays a pivotal role in viral replication and is a potential target for anti-SARS therapy. There is a lack of structural or biochem. data on any coronavirus polymerase. To provide insights into the structure and function of SARS-CoV RdRp, we have located its conserved motifs that are shared by all RdRps, and built a three-dimensional model of the catalytic domain. The structural model permits us to discuss the potential functional roles of the conserved motifs and residues in replication and their potential interactions with inhibitors of related enzymes. We predict the following important structural attributes for potential anti-SARS-CoV RdRp nucleotide analog inhibitors: hydrogen-bonding capability for the 2' and 3' groups of the sugar ring and C3' endo sugar puckering, and the absence of a hydrophobic binding pocket for non-nucleoside analog inhibitors similar to those obsd. in hepatitis C virus RdRp and human immunodeficiency virus type 1 reverse transcriptase. We propose that the clin. obsd. resistance of SARS to ribavirin is probably due to perturbation of the conserved motif A that controls rNTP binding and fidelity of polymn. Our results suggest that designing anti-SARS therapies can benefit from successful experiences in design of other antiviral drugs. This work should also provide guidance for future biochem. expts.
- 8De Clercq, E.; Li, G. Approved Antiviral Drugs over the Past 50 Years. Clin. Microbiol. Rev. 2016, 29 (3), 695– 747, DOI: 10.1128/CMR.00102-15Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2s%252Fos12htA%253D%253D&md5=1d25d961c1c6a72d6bc4634041bbb6feApproved Antiviral Drugs over the Past 50 YearsDe Clercq Erik; Li GuangdiClinical microbiology reviews (2016), 29 (3), 695-747 ISSN:.Since the first antiviral drug, idoxuridine, was approved in 1963, 90 antiviral drugs categorized into 13 functional groups have been formally approved for the treatment of the following 9 human infectious diseases: (i) HIV infections (protease inhibitors, integrase inhibitors, entry inhibitors, nucleoside reverse transcriptase inhibitors, nonnucleoside reverse transcriptase inhibitors, and acyclic nucleoside phosphonate analogues), (ii) hepatitis B virus (HBV) infections (lamivudine, interferons, nucleoside analogues, and acyclic nucleoside phosphonate analogues), (iii) hepatitis C virus (HCV) infections (ribavirin, interferons, NS3/4A protease inhibitors, NS5A inhibitors, and NS5B polymerase inhibitors), (iv) herpesvirus infections (5-substituted 2'-deoxyuridine analogues, entry inhibitors, nucleoside analogues, pyrophosphate analogues, and acyclic guanosine analogues), (v) influenza virus infections (ribavirin, matrix 2 protein inhibitors, RNA polymerase inhibitors, and neuraminidase inhibitors), (vi) human cytomegalovirus infections (acyclic guanosine analogues, acyclic nucleoside phosphonate analogues, pyrophosphate analogues, and oligonucleotides), (vii) varicella-zoster virus infections (acyclic guanosine analogues, nucleoside analogues, 5-substituted 2'-deoxyuridine analogues, and antibodies), (viii) respiratory syncytial virus infections (ribavirin and antibodies), and (ix) external anogenital warts caused by human papillomavirus infections (imiquimod, sinecatechins, and podofilox). Here, we present for the first time a comprehensive overview of antiviral drugs approved over the past 50 years, shedding light on the development of effective antiviral treatments against current and emerging infectious diseases worldwide.
- 9Kanters, S.; Socias, M. E.; Paton, N. I.; Vitoria, M.; Doherty, M.; Ayers, D.; Popoff, E.; Chan, K.; Cooper, D. A.; Wiens, M. O.; Calmy, A.; Ford, N.; Nsanzimana, S.; Mills, E. J. Comparative efficacy and safety of second-line antiretroviral therapy for treatment of HIV/AIDS: A systematic review and network meta-analysis. Lancet HIV 2017, 4 (10), e433– e441, DOI: 10.1016/S2352-3018(17)30109-1Google ScholarThere is no corresponding record for this reference.
- 10Deeks, E. D. Darunavir: a review of its use in the management of HIV-1 infection. Drugs 2014, 74 (1), 99– 125, DOI: 10.1007/s40265-013-0159-3Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXptFajuw%253D%253D&md5=b5f8e515fd0c0ce64d97b0c126c18ce4Darunavir: A Review of Its Use in the Management of HIV-1 InfectionDeeks, Emma D.Drugs (2014), 74 (1), 99-125CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. The latest HIV-1 protease inhibitor (PI) darunavir (Prezista®) has a high genetic barrier to resistance development and is active against wild-type HIV and HIV strains no longer susceptible to some older PIs. Ritonavir-boosted darunavir, as a component of antiretroviral therapy (ART), is indicated for the treatment of HIV-1 infection in adult and paediatric patients (aged ≥3 years), with or without treatment experience (details vary depending on region of approval). Several open-label or partially-blinded trials have evaluated the efficacy of ritonavir-boosted darunavir ART regimens for up to 192 wk in these settings. In treatment-naive adults, once-daily boosted darunavir was no less effective in establishing virol. suppression than once- or twice-daily boosted lopinavir, yet was more effective at maintaining suppression long term. Moreover, treatment-experienced adults with no darunavir resistance-assocd. mutations (RAMs) had no less effective viral load suppression with once-daily than with twice-daily boosted darunavir. In treatment-experienced adults, including some with multiple major PI RAMs, twice-daily boosted darunavir was more effective than twice-daily boosted lopinavir or boosted control PIs in reducing viral load, and provided virol. benefit as part of a salvage regimen in those with few remaining treatment options. Boosted darunavir also reduced viral load when administered once-daily in treatment-naive adolescents or twice-daily in treatment-experienced children and adolescents. Boosted darunavir is generally well tolerated, with gastrointestinal disturbances and lipid abnormalities among the most common tolerability issues. It has a lipid profile more favorable than that of boosted lopinavir in terms of total cholesterol and triglyceride changes and, when administered once daily, its lipid effects are generally similar to those of boosted atazanavir. Thus, boosted darunavir is a useful option for the ART regimens of adult and paediatric patients with HIV-1 infection.
- 11la Porte, C. J. Saquinavir, the pioneer antiretroviral protease inhibitor. Expert Opin. Drug Metab. Toxicol. 2009, 5 (10), 1313– 1322, DOI: 10.1517/17425250903273160Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFCgsrfM&md5=337b9225b09c6cabc0a87f41272b1edeSaquinavir, the pioneer antiretroviral protease inhibitorLa Porte, Charles J. L.Expert Opinion on Drug Metabolism & Toxicology (2009), 5 (10), 1313-1322CODEN: EODMAP; ISSN:1742-5255. (Informa Healthcare)A review. Background: The treatment of HIV infection underwent a major change in 1995 when saquinavir was the first protease inhibitor introduced into the market. This drug made the use of combination therapy in the treatment of HIV possible and increased the success rate of treatment. Objective: This article will review recent literature on saquinavir to define its current role in HIV treatment, among the newer antiretroviral drugs. Methods: Scientific literature and conference presentations were evaluated for relevant information pertaining to saquinavir. Results/conclusions: Although underused, saquinavir has good efficacy and tolerability when compared to other protease inhibitors. The film-coated tablet formulation improved pill burden. Saquinavir still has potential in the treatment of adults, children and pregnant women.
- 12Heo, Y. A.; Deeks, E. D. Sofosbuvir/Velpatasvir/Voxilaprevir: A Review in Chronic Hepatitis C. Drugs 2018, 78 (5), 577– 587, DOI: 10.1007/s40265-018-0895-5Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkslGjt7k%253D&md5=d4557e43a321ca3a9a30d996ac5f77baSofosbuvir/Velpatasvir/Voxilaprevir: A Review in Chronic Hepatitis CHeo, Young-A.; Deeks, Emma D.Drugs (2018), 78 (5), 577-587CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A fixed-dose combination of the hepatitis C virus (HCV) NS5B polymerase inhibitor sofosbuvir, the HCV NS5A inhibitor velpatasvir and the HCV NS3/4A protease inhibitor voxilaprevir (sofosbuvir/velpatasvir/voxilaprevir; Vosevi) is approved in the EU for the treatment of chronic HCV genotype 1, 2, 3, 4, 5 or 6 infection in adults. In the phase III POLARIS trials, in patients who had HCV genotype 1-6 infection with or without compensated cirrhosis, overall rates of sustained virol. response at 12 wk post-treatment (SVR12) with sofosbuvir/velpatasvir/voxilaprevir were high after 8 wk of treatment in direct-acting antiviral (DAA)-naive patients and 12 wk of treatment in DAA-experienced patients. However, 8 wk of sofosbuvir/velpatasvir/voxilaprevir was inferior to 12 wk of sofosbuvir/velpatasvir in cirrhotic or non-cirrhotic DAA-naive patients with HCV genotype 1, 2, 4, 5 or 6 infection and non-cirrhotic DAA-naive patients with HCV genotype 3 infection, mostly due to an insufficient treatment period. Sofosbuvir/velpatasvir/voxilaprevir was generally well tolerated, with most adverse events being of mild or moderate intensity. The most common adverse events included headache, fatigue, nausea and diarrhoea. In conclusion, sofosbuvir/velpatasvir/voxilaprevir is an important and effective option for the treatment of HCV genotype 1-6 infection in adults, esp. those who have previously failed a DAA therapy with or without an HCV NS5A inhibitor.
- 13Sanford, M. Simeprevir: a review of its use in patients with chronic hepatitis C virus infection. Drugs 2015, 75 (2), 183– 196, DOI: 10.1007/s40265-014-0341-2Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVCnt7k%253D&md5=20ef9a811518a120a68eb79241f0d735Simeprevir: A Review of Its Use in Patients with Chronic Hepatitis C Virus InfectionSanford, MarkDrugs (2015), 75 (2), 183-196CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. Simeprevir (Olysio; Galexos; Sovriad) is an orally-administered NS3/4A protease inhibitor for use in combined drug regimens against chronic hepatitis C virus (HCV) infection. This article reviews studies relevant to the EU simeprevir label. In proof-of-concept studies, simeprevir had potent antiviral activity against all HCV genotypes, except genotype 3. In trials in patients with chronic HCV genotype 1 infection, week-12 sustained virol. response (SVR12) rates in treatment-naive patients and prior relapsers were significantly higher with simeprevir plus peginterferon-α/ribavirin (PR) [79-89 %] than with placebo plus PR (36-62 %). In prior partial/null responders, the SVR12 rate with simeprevir plus PR (54 %) was noninferior to that with telaprevir plus PR (55 %). Simeprevir plus PR was also efficacious in patients with HCV genotype 1/HIV-1 co-infection. In prior null responders without severe liver fibrosis (cohort 1) and treatment-naive patients with severe liver fibrosis (cohort 2) treated with simeprevir plus sofosbuvir, the SVR12 rate for the two cohorts combined was 92 %. In patients with chronic HCV genotype 4 infection, the SVR12 rates with simeprevir plus PR were 83, 87 and 40 % in treatment-naive patients, prior relapsers and prior null responders, resp. Grade 3-4 adverse event, serious adverse event and treatment withdrawal rates with simeprevir plus PR were similar to those with placebo plus PR. Skin rashes with simeprevir were mostly mild or moderate; serious photosensitivity reactions occur, but are rare. Simeprevir is efficacious and generally well tolerated in patients with chronic HCV genotypes 1 and 4 infection. Studies of simeprevir in interferon-free regimens and in other subpopulations with HCV infections will be of interest.
- 14Chopp, S.; Vanderwall, R.; Hult, A.; Klepser, M. Simeprevir and sofosbuvir for treatment of hepatitis C infection. Am. J. Health-Syst. Pharm. 2015, 72 (17), 1445– 1455, DOI: 10.2146/ajhp140290Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFGgt7k%253D&md5=b8e2a8a629e121eeba1b7bb37a5fa5e6Simeprevir and sofosbuvir for treatment of hepatitis C infectionChopp, Shelby; Vanderwall, Rebecca; Hult, Amanda; Klepser, MichaelAmerican Journal of Health-System Pharmacy (2015), 72 (17), 1445-1455CODEN: AHSPEK; ISSN:1079-2082. (American Society of Health-System Pharmacists)Purpose. The pharmacol., pharmacokinetics, efficacy, safety, costs, and place in therapy of simeprevir and sofosbuvir in the management of hepatitis C virus (HCV) infection are reviewed. Summary. Sofosbuvir and simeprevir are classified as direct-acting agents because they target specific proteins essential to the replication of HCV. Phase III trials demonstrated that simeprevir in combination with peginterferon alfa and ribavirin was superior to placebo combined with peginterferon alfa and ribavirin in achieving a sustained virol. response in both treatment-naive patients and patients who relapsed after treatment with peginterferon alfa-2a or alfa-2b and ribavirin. Q80K polymorphism substantially decreases the efficacy of simeprevir. Clin. trials revealed that sofosbuvir in combination with ribavirin was superior to peginterferon plus ribavirin against HCV genotype 2 infection and as effective as peginterferon plus ribavirin against HCV genotype 3 infection. These findings were significant because they demonstrated the effectiveness of an anti-HCV regimen that did not include peginterferon alfa. Sofosbuvir has much better adverse-effect and drug interaction profiles than previous hepatitis C antiviral agents. Both simeprevir and sofosbuvir are approved for the treatment of chronic hepatitis C in combination with other antiviral medications. Simeprevir has been approved specifically for patients infected with HCV genotype 1 with compensated liver disease (including cirrhosis) in combination with peginterferon alfa-2a or alfa-2b and ribavirin. Sofosbuvir has shown efficacy in HCV genotypes 1-4. Conclusion. Simeprevir and sofasbuvir have advantages in response rates and convenient dosage forms and frequency compared with other HCV treatments; however, they are more expensive than previous HCV therapies.
- 15Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; Duan, Y.; Yu, J.; Wang, L.; Yang, K.; Liu, F.; Jiang, R.; Yang, X.; You, T.; Liu, X.; Yang, X.; Bai, F.; Liu, H.; Liu, X.; Guddat, L. W.; Xu, W.; Xiao, G.; Qin, C.; Shi, Z.; Jiang, H.; Rao, Z.; Yang, H. Structure of M(pro) from COVID-19 virus and discovery of its inhibitors. Nature 2020, 582, 289– 293, DOI: 10.1038/s41586-020-2223-yGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVyhsrrO&md5=b84f350fe9ce1109485df6caf814ba82Structure of Mpro from SARS-CoV-2 and discovery of its inhibitorsJin, Zhenming; Du, Xiaoyu; Xu, Yechun; Deng, Yongqiang; Liu, Meiqin; Zhao, Yao; Zhang, Bing; Li, Xiaofeng; Zhang, Leike; Peng, Chao; Duan, Yinkai; Yu, Jing; Wang, Lin; Yang, Kailin; Liu, Fengjiang; Jiang, Rendi; Yang, Xinglou; You, Tian; Liu, Xiaoce; Yang, Xiuna; Bai, Fang; Liu, Hong; Liu, Xiang; Guddat, Luke W.; Xu, Wenqing; Xiao, Gengfu; Qin, Chengfeng; Shi, Zhengli; Jiang, Hualiang; Rao, Zihe; Yang, HaitaoNature (London, United Kingdom) (2020), 582 (7811), 289-293CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new coronavirus, known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the etiol. agent responsible for the 2019-2020 viral pneumonia outbreak of coronavirus disease 2019 (COVID-19). Currently, there are no targeted therapeutic agents for the treatment of this disease, and effective treatment options remain very limited. Here, we describe the results of a program that aimed to rapidly discover lead compds. for clin. use, by combining structure-assisted drug design, virtual drug screening and high-throughput screening. This program focused on identifying drug leads that target main protease (Mpro) of SARS-CoV-2: Mpro is a key enzyme of coronaviruses and has a pivotal role in mediating viral replication and transcription, making it an attractive drug target for SARS-CoV-2. We identified a mechanism-based inhibitor (N3) by computer-aided drug design, and then detd. the crystal structure of Mpro of SARS-CoV-2 in complex with this compd. Through a combination of structure-based virtual and high-throughput screening, we assayed more than 10,000 compds.-including approved drugs, drug candidates in clin. trials and other pharmacol. active compds.-as inhibitors of Mpro. Six of these compds. inhibited Mpro, showing half-maximal inhibitory concn. values that ranged from 0.67 to 21.4μM. One of these compds. (ebselen) also exhibited promising antiviral activity in cell-based assays. Our results demonstrate the efficacy of our screening strategy, which can lead to the rapid discovery of drug leads with clin. potential in response to new infectious diseases for which no specific drugs or vaccines are available.
- 16Dai, W.; Zhang, B.; Jiang, X. M.; Su, H.; Li, J.; Zhao, Y.; Xie, X.; Jin, Z.; Peng, J.; Liu, F.; Li, C.; Li, Y.; Bai, F.; Wang, H.; Cheng, X.; Cen, X.; Hu, S.; Yang, X.; Wang, J.; Liu, X.; Xiao, G.; Jiang, H.; Rao, Z.; Zhang, L. K.; Xu, Y.; Yang, H.; Liu, H. Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease. Science 2020, 368 (6497), 1331– 1335, DOI: 10.1126/science.abb4489Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1alsrzL&md5=ec87c340410edb919d2584a2b4d33e1aStructure-based design of antiviral drug candidates targeting the SARS-CoV-2 main proteaseDai, Wenhao; Zhang, Bing; Jiang, Xia-Ming; Su, Haixia; Li, Jian; Zhao, Yao; Xie, Xiong; Jin, Zhenming; Peng, Jingjing; Liu, Fengjiang; Li, Chunpu; Li, You; Bai, Fang; Wang, Haofeng; Cheng, Xi; Cen, Xiaobo; Hu, Shulei; Yang, Xiuna; Wang, Jiang; Liu, Xiang; Xiao, Gengfu; Jiang, Hualiang; Rao, Zihe; Zhang, Lei-Ke; Xu, Yechun; Yang, Haitao; Liu, HongScience (Washington, DC, United States) (2020), 368 (6497), 1331-1335CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is the etiol. agent responsible for the global COVID-19 (coronavirus disease 2019) outbreak. The main protease of SARS-CoV-2, Mpro, is a key enzyme that plays a pivotal role in mediating viral replication and transcription. We designed and synthesized two lead compds. (11a and 11b) targeting Mpro. Both exhibited excellent inhibitory activity and potent anti-SARS-CoV-2 infection activity. The x-ray crystal structures of SARS-CoV-2 Mpro in complex with 11a or 11b, both detd. at a resoln. of 1.5 angstroms, showed that the aldehyde groups of 11a and 11b are covalently bound to cysteine 145 of Mpro. Both compds. showed good pharmacokinetic properties in vivo, and 11a also exhibited low toxicity, which suggests that these compds. are promising drug candidates.
- 17Vuong, W.; Khan, M. B.; Fischer, C.; Arutyunova, E.; Lamer, T.; Shields, J.; Saffran, H. A.; McKay, R. T.; van Belkum, M. J.; Joyce, M. A.; Young, H. S.; Tyrrell, D. L.; Vederas, J. C.; Lemieux, M. J. Feline coronavirus drug inhibits the main protease of SARS-CoV-2 and blocks virus replication. Nat. Commun. 2020, 11 (1), 4282, DOI: 10.1038/s41467-020-18096-2Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslWqtbzN&md5=c06d2dc9a65ae289505fe120d41d8885Feline coronavirus drug inhibits the main protease of SARS-CoV-2 and blocks virus replicationVuong, Wayne; Khan, Muhammad Bashir; Fischer, Conrad; Arutyunova, Elena; Lamer, Tess; Shields, Justin; Saffran, Holly A.; McKay, Ryan T.; van Belkum, Marco J.; Joyce, Michael A.; Young, Howard S.; Tyrrell, D. Lorne; Vederas, John C.; Lemieux, M. JoanneNature Communications (2020), 11 (1), 4282CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The main protease, Mpro (or 3CLpro) in SARS-CoV-2 is a viable drug target because of its essential role in the cleavage of the virus polypeptide. Feline infectious peritonitis, a fatal coronavirus infection in cats, was successfully treated previously with a prodrug GC376, a dipeptide-based protease inhibitor. We show the prodrug and its parent GC373, are effective inhibitors of the Mpro from both SARS-CoV and SARS-CoV-2 with IC50 values in the nanomolar range. Crystal structures of SARS-CoV-2 Mpro with these inhibitors have a covalent modification of the nucleophilic Cys145. NMR anal. reveals that inhibition proceeds via reversible formation of a hemithioacetal. GC373 and GC376 are potent inhibitors of SARS-CoV-2 replication in cell culture. They are strong drug candidates for the treatment of human coronavirus infections because they have already been successful in animals. The work here lays the framework for their use in human trials for the treatment of COVID-19.
- 18Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved alpha-ketoamide inhibitors. Science 2020, 368 (6489), 409– 412, DOI: 10.1126/science.abb3405Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnslKrtL8%253D&md5=9ac417c20f54c3327f9de9088b512d52Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitorsZhang, Linlin; Lin, Daizong; Sun, Xinyuanyuan; Curth, Ute; Drosten, Christian; Sauerhering, Lucie; Becker, Stephan; Rox, Katharina; Hilgenfeld, RolfScience (Washington, DC, United States) (2020), 368 (6489), 409-412CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is a global health emergency. An attractive drug target among coronaviruses is the main protease (Mpro, also called 3CLpro) because of its essential role in processing the polyproteins that are translated from the viral RNA. We report the x-ray structures of the unliganded SARS-CoV-2 Mpro and its complex with an α-ketoamide inhibitor. This was derived from a previously designed inhibitor but with the P3-P2 amide bond incorporated into a pyridone ring to enhance the half-life of the compd. in plasma. On the basis of the unliganded structure, we developed the lead compd. into a potent inhibitor of the SARS-CoV-2 Mpro. The pharmacokinetic characterization of the optimized inhibitor reveals a pronounced lung tropism and suitability for administration by the inhalative route.
- 19Jin, Z.; Zhao, Y.; Sun, Y.; Zhang, B.; Wang, H.; Wu, Y.; Zhu, Y.; Zhu, C.; Hu, T.; Du, X.; Duan, Y.; Yu, J.; Yang, X.; Yang, X.; Yang, K.; Liu, X.; Guddat, L. W.; Xiao, G.; Zhang, L.; Yang, H.; Rao, Z. Structural basis for the inhibition of SARS-CoV-2 main protease by antineoplastic drug carmofur. Nat. Struct. Mol. Biol. 2020, 27 (6), 529– 532, DOI: 10.1038/s41594-020-0440-6Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovVOrtLY%253D&md5=28c7bed4e4154935e6909a53015bf64cStructural basis for the inhibition of SARS-CoV-2 main protease by antineoplastic drug carmofurJin, Zhenming; Zhao, Yao; Sun, Yuan; Zhang, Bing; Wang, Haofeng; Wu, Yan; Zhu, Yan; Zhu, Chen; Hu, Tianyu; Du, Xiaoyu; Duan, Yinkai; Yu, Jing; Yang, Xiaobao; Yang, Xiuna; Yang, Kailin; Liu, Xiang; Guddat, Luke W.; Xiao, Gengfu; Zhang, Leike; Yang, Haitao; Rao, ZiheNature Structural & Molecular Biology (2020), 27 (6), 529-532CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)Abstr.: The antineoplastic drug carmofur is shown to inhibit the SARS-CoV-2 main protease (Mpro). Here, the X-ray crystal structure of Mpro in complex with carmofur reveals that the carbonyl reactive group of carmofur is covalently bound to catalytic Cys145, whereas its fatty acid tail occupies the hydrophobic S2 subsite. Carmofur inhibits viral replication in cells (EC50 = 24.30μM) and is a promising lead compd. to develop new antiviral treatment for COVID-19.
- 20Fu, L.; Ye, F.; Feng, Y.; Yu, F.; Wang, Q.; Wu, Y.; Zhao, C.; Sun, H.; Huang, B.; Niu, P.; Song, H.; Shi, Y.; Li, X.; Tan, W.; Qi, J.; Gao, G. F. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat. Commun. 2020, 11 (1), 4417, DOI: 10.1038/s41467-020-18233-xGoogle Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVSmsr3F&md5=273b11d16241499f15103885be398b23Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main proteaseFu, Lifeng; Ye, Fei; Feng, Yong; Yu, Feng; Wang, Qisheng; Wu, Yan; Zhao, Cheng; Sun, Huan; Huang, Baoying; Niu, Peihua; Song, Hao; Shi, Yi; Li, Xuebing; Tan, Wenjie; Qi, Jianxun; Gao, George FuNature Communications (2020), 11 (1), 4417CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: COVID-19 was declared a pandemic on March 11 by WHO, due to its great threat to global public health. The coronavirus main protease (Mpro, also called 3CLpro) is essential for processing and maturation of the viral polyprotein, therefore recognized as an attractive drug target. Here we show that a clin. approved anti-HCV drug, Boceprevir, and a pre-clin. inhibitor against feline infectious peritonitis (corona) virus (FIPV), GC376, both efficaciously inhibit SARS-CoV-2 in Vero cells by targeting Mpro. Moreover, combined application of GC376 with Remdesivir, a nucleotide analog that inhibits viral RNA dependent RNA polymerase (RdRp), results in sterilizing additive effect. Further structural anal. reveals binding of both inhibitors to the catalytically active side of SARS-CoV-2 protease Mpro as main mechanism of inhibition. Our findings may provide crit. information for the optimization and design of more potent inhibitors against the emerging SARS-CoV-2 virus.
- 21Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 2004, 47 (7), 1739– 1749, DOI: 10.1021/jm0306430Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit74%253D&md5=8cc2f0022318b12dd972e9c493375bf9Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessment of docking accuracyFriesner, Richard A.; Banks, Jay L.; Murphy, Robert B.; Halgren, Thomas A.; Klicic, Jasna J.; Mainz, Daniel T.; Repasky, Matthew P.; Knoll, Eric H.; Shelley, Mee; Perry, Jason K.; Shaw, David E.; Francis, Perry; Shenkin, Peter S.Journal of Medicinal Chemistry (2004), 47 (7), 1739-1749CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Unlike other methods for docking ligands to the rigid 3D structure of a known protein receptor, Glide approximates a complete systematic search of the conformational, orientational, and positional space of the docked ligand. In this search, an initial rough positioning and scoring phase that dramatically narrows the search space is followed by torsionally flexible energy optimization on an OPLS-AA nonbonded potential grid for a few hundred surviving candidate poses. The very best candidates are further refined via a Monte Carlo sampling of pose conformation; in some cases, this is crucial to obtaining an accurate docked pose. Selection of the best docked pose uses a model energy function that combines empirical and force-field-based terms. Docking accuracy is assessed by redocking ligands from 282 cocrystd. PDB complexes starting from conformationally optimized ligand geometries that bear no memory of the correctly docked pose. Errors in geometry for the top-ranked pose are less than 1 Å in nearly half of the cases and are greater than 2 Å in only about one-third of them. Comparisons to published data on rms deviations show that Glide is nearly twice as accurate as GOLD and more than twice as accurate as FlexX for ligands having up to 20 rotatable bonds. Glide is also found to be more accurate than the recently described Surflex method.
- 22Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. J. Med. Chem. 2004, 47 (7), 1750– 1759, DOI: 10.1021/jm030644sGoogle Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit78%253D&md5=33d68dd968e65626b449df61e44e37beGlide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screeningHalgren, Thomas A.; Murphy, Robert B.; Friesner, Richard A.; Beard, Hege S.; Frye, Leah L.; Pollard, W. Thomas; Banks, Jay L.Journal of Medicinal Chemistry (2004), 47 (7), 1750-1759CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Glide's ability to identify active compds. in a database screen is characterized by applying Glide to a diverse set of nine protein receptors. In many cases, two, or even three, protein sites are employed to probe the sensitivity of the results to the site geometry. To make the database screens as realistic as possible, the screens use sets of "druglike" decoy ligands that have been selected to be representative of what we believe is likely to be found in the compd. collection of a pharmaceutical or biotechnol. company. Results are presented for releases 1.8, 2.0, and 2.5 of Glide. The comparisons show that av. measures for both "early" and "global" enrichment for Glide 2.5 are 3 times higher than for Glide 1.8 and more than 2 times higher than for Glide 2.0 because of better results for the least well-handled screens. This improvement in enrichment stems largely from the better balance of the more widely parametrized GlideScore 2.5 function and the inclusion of terms that penalize ligand-protein interactions that violate established principles of phys. chem., particularly as it concerns the exposure to solvent of charged protein and ligand groups. Comparisons to results for the thymidine kinase and estrogen receptors published by Rognan and co-workers (J. Med. Chem. 2000, 43, 4759-4767) show that Glide 2.5 performs better than GOLD 1.1, FlexX 1.8, or DOCK 4.01.
- 23Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J. Med. Chem. 2006, 49 (21), 6177– 6196, DOI: 10.1021/jm051256oGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpvVGmurg%253D&md5=ea428c82ead0d8c27f8c1a7b694a1edfExtra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 24Amporndanai, K.; Meng, X.; Shang, W.; Jin, Z.; Rogers, M.; Zhao, Y.; Rao, Z.; Liu, Z. J.; Yang, H.; Zhang, L.; O’Neill, P. M.; Samar Hasnain, S. Inhibition mechanism of SARS-CoV-2 main protease by ebselen and its derivatives. Nat. Commun. 2021, 12 (1), 3061, DOI: 10.1038/s41467-021-23313-7Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtFyntbvK&md5=08fe8105502a0bd2aec4879d78280716Inhibition mechanism of SARS-CoV-2 main protease by ebselen and its derivativesAmporndanai, Kangsa; Meng, Xiaoli; Shang, Weijuan; Jin, Zhenmig; Rogers, Michael; Zhao, Yao; Rao, Zihe; Liu, Zhi-Jie; Yang, Haitao; Zhang, Leike; O'Neill, Paul M.; Samar Hasnain, S.Nature Communications (2021), 12 (1), 3061CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: The SARS-CoV-2 pandemic has triggered global efforts to develop therapeutics. The main protease of SARS-CoV-2 (Mpro), crit. for viral replication, is a key target for therapeutic development. An organoselenium drug called ebselen has been demonstrated to have potent Mpro inhibition and antiviral activity. We have examd. the binding modes of ebselen and its deriv. in Mpro via high resoln. co-crystallog. and investigated their chem. reactivity via mass spectrometry. Stronger Mpro inhibition than ebselen and potent ability to rescue infected cells were obsd. for a no. of derivs. A free selenium atom bound with cysteine of catalytic dyad has been revealed in crystallog. structures of Mpro with ebselen and MR6-31-2 suggesting hydrolysis of the enzyme bound organoselenium covalent adduct and formation of a phenolic byproduct, confirmed by mass spectrometry. The target engagement with selenation mechanism of inhibition suggests wider therapeutic applications of these compds. against SARS-CoV-2 and other zoonotic beta-corona viruses.
- 25Muramatsu, T.; Takemoto, C.; Kim, Y. T.; Wang, H.; Nishii, W.; Terada, T.; Shirouzu, M.; Yokoyama, S. SARS-CoV 3CL protease cleaves its C-terminal autoprocessing site by novel subsite cooperativity. Proc. Natl. Acad. Sci. U. S. A. 2016, 113 (46), 12997– 13002, DOI: 10.1073/pnas.1601327113Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslKnu7%252FP&md5=f82749f292da1066e3eb4717dcbc66f2SARS-CoV 3CL protease cleaves its C-terminal autoprocessing site by novel subsite cooperativityMuramatsu, Tomonari; Takemoto, Chie; Kim, Yong-Tae; Wang, Hongfei; Nishii, Wataru; Terada, Takaho; Shirouzu, Mikako; Yokoyama, ShigeyukiProceedings of the National Academy of Sciences of the United States of America (2016), 113 (46), 12997-13002CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The 3C-like protease (3CLpro) of severe acute respiratory syndrome coronavirus (SARS-CoV) cleaves 11 sites in the polyproteins, including its own N- and C-terminal autoprocessing sites, by recognizing P4-P1 and P1'. In this study, we detd. the crystal structure of 3CLpro with the C-terminal prosequence and the catalytic-site C145A mutation, in which the enzyme binds the C-terminal prosequence of another mol. Surprisingly, Phe at the P3' position [Phe(P3')] is snugly accommodated in the S3' pocket. Mutations of Phe(P3') impaired the C-terminal autoprocessing, but did not affect N-terminal autoprocessing. This difference was ascribed to the P2 residue, Phe(P2) and Leu(P2), in the C- and N-terminal sites, as follows. The S3' subsite is formed by Phe(P2)-induced conformational changes of 3CLpro and the direct involvement of Phe(P2) itself. In contrast, the N-terminal prosequence with Leu(P2) does not cause such conformational changes for the S3' subsite formation. In fact, the mutation of Phe(P2) to Leu in the C-terminal autoprocessing site abolishes the dependence on Phe(P3'). These mechanisms explain why Phe is required at the P3' position when the P2 position is occupied by Phe rather than Leu, which reveals a type of subsite cooperativity. Moreover, the peptide consisting of P4-P1 with Leu(P2) inhibits protease activity, whereas that with Phe(P2) exhibits a much smaller inhibitory effect, because Phe(P3') is missing. Thus, this subsite cooperativity likely exists to avoid the autoinhibition of the enzyme by its mature C-terminal sequence, and to retain the efficient C-terminal autoprocessing by the use of Phe(P2).
- 26Sarma, B. K.; Mugesh, G. Antioxidant activity of the anti-inflammatory compound ebselen: a reversible cyclization pathway via selenenic and seleninic acid intermediates. Chem. - Eur. J. 2008, 14 (34), 10603– 10614, DOI: 10.1002/chem.200801258Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2rsLfI&md5=6a82b84de2f8fddc7f2ac90cd62e0b15Antioxidant activity of the anti-inflammatory compound ebselen: a reversible cyclization pathway via selenenic and seleninic acid intermediatesSarma, Bani Kanta; Mugesh, GovindasamyChemistry - A European Journal (2008), 14 (34), 10603-10614CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)A revised mechanism that accounts for the glutathione peroxidase (GPx)-like catalytic activity of the organoselenium compd. ebselen (2-phenyl-1,2-benzisoselenazol-3(2H)-one) is described. The reaction of ebselen with H2O2 yields seleninic acid 2-PhNHC(O)C6H4SeO2H as the only oxidized product. The X- ray crystal structure of the seleninic acid shows that the Se atom is involved in a noncovalent interaction with the carbonyl O atom. In the presence of excess PhSH, the Se-N bond in ebselen is readily cleaved by the thiol to produce the corresponding selenenyl sulfide 2-PhNHC(O)C6H4SeSPh. The selenenyl sulfide thus produced undergoes a disproportionation in the presence of H2O2 to produce the diselenide, which upon reaction with H2O2, produces a mixt. of selenenic and seleninic acids. The addn. of thiol to the mixt. contg. selenenic and seleninic acids gives the selenenyl sulfide. When the concn. of the thiol is relatively low in the reaction mixt., the selenenic acid undergoes a rapid cyclization to produce ebselen. The seleninic acid, however, reacts with the diselenide to produce ebselen as the final product. DFT calcns. show that the cyclization of selenenic acids to the corresponding selenenyl amides is more favored than that of sulfenic acids to the corresponding sulfenyl amides. The regeneration of ebselen under a variety of conditions protects the Se moiety from irreversible inactivation, which may be responsible for the biol. activities of ebselen.
- 27Haenen, G. R.; De Rooij, B. M.; Vermeulen, N. P.; Bast, A. Mechanism of the reaction of ebselen with endogenous thiols: Dihydrolipoate is a better cofactor than glutathione in the peroxidase activity of ebselen. Mol. Pharmacol. 1990, 37 (3), 412– 422Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXhvFWlsrg%253D&md5=ce9215a47abfe5cd650595c67a39c9c7Mechanism of the reaction of ebselen with endogenous thiols: dihydrolipoate is a better cofactor than glutathione in the peroxidase activity of ebselenHaenen, Guido R. M. M.; De Rooij, Ben M.; Vermeulen, Nico P. E.; Bast, AaltMolecular Pharmacology (1990), 37 (3), 412-22CODEN: MOPMA3; ISSN:0026-895X.The therapeutic effect of ebselen has been linked to its peroxidase activity. In the present study, the peroxidase activity of ebselen toward H2O2 with the endogenous thiols GSH and dihydrolipoate [L(SH)2] as cofactors was detd. When GSH was used, peroxide removal was described by a ter uni ping pong mechanism with Dalziel coeffs. for GSH and H2O2 of 0.165 and 0.081 mM min, resp. When L(SH)2 was used, peroxidase activity was independent of the concn. of L(SH)2, in the concn. range studied (5 μM to 2 mM) and peroxide removal was only dependent on the concn. of H2O2 and ebselen, with the second-order rate const. being 12.3 mM-1 min-1. To elucidate the difference between GSH and L(SH)2, the mol. mechanism of the peroxidase activity of ebselen was investigated, using UV spectrophotometry, HPLC, 77Se NMR, and mass spectrometry. GSH was found to react quickly with ebselen to give a selenenyl sulfide, an adduct of GSH to ebselen. Subsequently, the GSH-selenenyl sulfide is converted into the diselenide of ebselen. Finally the diselenide reacts with a peroxide and ebselen is regenerated. The formation by GSH of the diselenide from the GSH-selenenyl sulfide of ebselen is slow and linearly dependent on the concn. of free thiol; however, no net consumption of GSH was obsd. Furthermore, it is likely that a selenol is an intermediate in diselenide formation. After reaction between ebselen and L(SH)2 the diselenide of ebselen was immediately detected. The fast formation of the diselenide with L(SH)2 vs. the slow formation of the diselenide with GSH accounts for our observation that L(SH)2 is a better cofactor than GSH in the peroxidase activity of ebselen. These results suggest that the interaction between ebselen and L(SH)2 might be of major importance in the mechanism by which ebselen exerts its therapeutic effect.
- 28Wagner, G.; Schuch, G.; Akerboom, T. P.; Sies, H. Transport of ebselen in plasma and its transfer to binding sites in the hepatocyte. Biochem. Pharmacol. 1994, 48 (6), 1137– 1144, DOI: 10.1016/0006-2952(94)90150-3Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXmt1WjsL8%253D&md5=78392c5dbfaa4f9e7a5476146b852a55Transport of ebselen in plasma and its transfer to binding sites in the hepatocyteWagner, Gunter; Schuch, Gunter; Akerboom, Theodorus P. M.; Sies, HelmutBiochemical Pharmacology (1994), 48 (6), 1137-44CODEN: BCPCA6; ISSN:0006-2952.In vivo transport in plasma and in vitro transfer of ebselen to binding sits in the hepatocyte were studied. More than 90% of i.v. administered ebselen in mouse plasma is bound by selenium-sulfur bonds to reactive thiols in serum albumin. In in vitro expts. the uptake of [14C]-ebselen from a complex prepd. with bovine serum albumin (BSA) was detd. in isolated perfused rat liver. Radioactive ebselen metabolites were excreted into bile. In isolated hepatocytes, radioactivity was bound to all subcellular organelles. Ebselen is transferred from the BSA complex to membrane-assocd. proteins after reductive cleavage of the Se-S bond effected by endogenous protein thiols. In contrast, when proteins were sepd. by dialysis membranes, ebselen transfer from its BSA complex occurred only in the presence of externally added reductants. Among the physiol. reductants tested, ebselen release from the BSA complex was highest with glutathione (75%) and lowest with ascorbic acid (less than 10%). Quant. release of ebselen from its BSA complex was only achieved by the combined action of reductant, notably 2-mercaptoethanol, and guanidine thiocyanate, suggesting that ebselen interacts with proteins by covalent Se-S bonds as well as by ionic charge interactions.
- 29Singh, N.; Halliday, A. C.; Thomas, J. M.; Kuznetsova, O. V.; Baldwin, R.; Woon, E. C.; Aley, P. K.; Antoniadou, I.; Sharp, T.; Vasudevan, S. R.; Churchill, G. C. A safe lithium mimetic for bipolar disorder. Nat. Commun. 2013, 4, 1332, DOI: 10.1038/ncomms2320Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3s3nvVSqsw%253D%253D&md5=682e37608458f5d24a55405d07a0e5b4A safe lithium mimetic for bipolar disorderSingh Nisha; Halliday Amy C; Thomas Justyn M; Kuznetsova Olga V; Baldwin Rhiannon; Woon Esther C Y; Aley Parvinder K; Antoniadou Ivi; Sharp Trevor; Vasudevan Sridhar R; Churchill Grant CNature communications (2013), 4 (), 1332 ISSN:.Lithium is the most effective mood stabilizer for the treatment of bipolar disorder, but it is toxic at only twice the therapeutic dosage and has many undesirable side effects. It is likely that a small molecule could be found with lithium-like efficacy but without toxicity through target-based drug discovery; however, therapeutic target of lithium remains equivocal. Inositol monophosphatase is a possible target but no bioavailable inhibitors exist. Here we report that the antioxidant ebselen inhibits inositol monophosphatase and induces lithium-like effects on mouse behaviour, which are reversed with inositol, consistent with a mechanism involving inhibition of inositol recycling. Ebselen is part of the National Institutes of Health Clinical Collection, a chemical library of bioavailable drugs considered clinically safe but without proven use. Therefore, ebselen represents a lithium mimetic with the potential both to validate inositol monophosphatase inhibition as a treatment for bipolar disorder and to serve as a treatment itself.
- 30Johnson, T. W.; Dress, K. R.; Edwards, M. Using the Golden Triangle to optimize clearance and oral absorption. Bioorg. Med. Chem. Lett. 2009, 19 (19), 5560– 5564, DOI: 10.1016/j.bmcl.2009.08.045Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFaktbfE&md5=3cfed1288ec451eb4419f1db753ae4b4Using the Golden Triangle to optimize clearance and oral absorptionJohnson, Ted W.; Dress, Klaus R.; Edwards, MartinBioorganic & Medicinal Chemistry Letters (2009), 19 (19), 5560-5564CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The Golden Triangle is a visualization tool developed from in vitro permeability, in vitro clearance and computational data designed to aid medicinal chemists in achieving metabolically stable, permeable and potent drug candidates. Classifying compds. as permeable and stable and plotting mol. wt. (MW) vs. octanol:buffer (pH 7.4) distribution coeffs. (log D) or estd. octanol:buffer (pH 7.4) distribution coeffs. (elog D) reveals useful trends. Anal. of at least 2 orthogonal trends, such as permeability and clearance, can be extremely effective in balancing and optimizing multiple properties. In addn., mol. wt. and log D impact potency-efficiency calcns., allowing potency, clearance and permeability to be optimized simultaneously.
- 31Johnson, T. W.; Gallego, R. A.; Edwards, M. P. Lipophilic Efficiency as an Important Metric in Drug Design. J. Med. Chem. 2018, 61 (15), 6401– 6420, DOI: 10.1021/acs.jmedchem.8b00077Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXmsVahsbg%253D&md5=29f38880914fa75c33be3a640ace4309Lipophilic Efficiency as an Important Metric in Drug DesignJohnson, Ted W.; Gallego, Rebecca A.; Edwards, Martin P.Journal of Medicinal Chemistry (2018), 61 (15), 6401-6420CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Lipophilic efficiency (LipE) is an important metric that has been increasingly applied in drug discovery medicinal chem. lead optimization programs. In this perspective, using literature drug discovery examples, we discuss the concept of rigorously applying LipE to guide medicinal chem. lead optimization toward drug candidates with potential for superior in vivo efficacy and safety, esp. when guided by physiochem. property-based optimization (PPBO). Also highlighted are examples of small structural modifications such as addn. of single atoms, small functional groups, and cyclizations that produce large increases in LipE. Understanding the factors that may contribute to LipE changes through anal. of ligand-protein crystal structures and using structure-based drug design (SBDD) to increase LipE by design is also discussed. Herein we advocate for use of LipE anal. coupled with PPBO and SBDD as an efficient mechanism for drug design.
- 32Hopkins, A. L.; Keseru, G. M.; Leeson, P. D.; Rees, D. C.; Reynolds, C. H. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discovery 2014, 13 (2), 105– 121, DOI: 10.1038/nrd4163Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1SmtLk%253D&md5=344030e59f499180f79d311302a27532The role of ligand efficiency metrics in drug discoveryHopkins, Andrew L.; Keserue, Gyoergy M.; Leeson, Paul D.; Rees, David C.; Reynolds, Charles H.Nature Reviews Drug Discovery (2014), 13 (2), 105-121CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The judicious application of ligand or binding efficiency metrics, which quantify the mol. properties required to obtain binding affinity for a drug target, is gaining traction in the selection and optimization of fragments, hits and leads. Retrospective anal. of recently marketed oral drugs shows that they frequently have highly optimized ligand efficiency values for their targets. Optimizing ligand efficiency metrics based on both mol. mass and lipophilicity, when set in the context of the specific target, has the potential to ameliorate the inflation of these properties that has been obsd. in current medicinal chem. practice, and to increase the quality of drug candidates.
- 33Raymer, B.; Bhattacharya, S. K. Lead-like Drugs: A Perspective. J. Med. Chem. 2018, 61 (23), 10375– 10384, DOI: 10.1021/acs.jmedchem.8b00407Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtl2is73O&md5=e9fc17d69c7d671459fdaf77d6721829Lead-like Drugs: A PerspectiveRaymer, Brian; Bhattacharya, Samit K.Journal of Medicinal Chemistry (2018), 61 (23), 10375-10384CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Lead-like drugs, or drugs below mol. wt. 300, are an important and sometimes overlooked component of the current pharmacopeia and contemporary medicinal chem. practice. To examine the recent state-of-the-art in lead-like drug discovery, we surveyed recent drug approvals from 2011 to 2017 and top 200 prescribed medications, as well as provide case studies on recently approved lead-like drugs. Many of these recent drugs are close analogs of previously known drugs or natural substrates, with a key focus of their medicinal chem. optimization being the choice of a low mol. wt. starting point and maintaining low mol. wt. during the optimization. However, the identification of low mol. wt. starting points may be limited by the availability of suitable low mol. wt. screening sets. To increase the discovery rate of lead-like drugs, we suggest an increased focus on inclusion and prosecution of lead-like starting points in screening libraries.
- 34Weiss, M. M.; Dineen, T. A.; Marx, I. E.; Altmann, S.; Boezio, A.; Bregman, H.; Chu-Moyer, M.; DiMauro, E. F.; Feric Bojic, E.; Foti, R. S.; Gao, H.; Graceffa, R.; Gunaydin, H.; Guzman-Perez, A.; Huang, H.; Huang, L.; Jarosh, M.; Kornecook, T.; Kreiman, C. R.; Ligutti, J.; La, D. S.; Lin, M. J.; Liu, D.; Moyer, B. D.; Nguyen, H. N.; Peterson, E. A.; Rose, P. E.; Taborn, K.; Youngblood, B. D.; Yu, V.; Fremeau, R. T., Jr. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities. J. Med. Chem. 2017, 60 (14), 5969– 5989, DOI: 10.1021/acs.jmedchem.6b01851Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXktFOmtbw%253D&md5=6333a6b936e48f9d18cf9df10ea12853Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic LiabilitiesWeiss, Matthew M.; Dineen, Thomas A.; Marx, Isaac E.; Altmann, Steven; Boezio, Alessandro; Bregman, Howard; Chu-Moyer, Margaret; DiMauro, Erin F.; Feric Bojic, Elma; Foti, Robert S.; Gao, Hua; Graceffa, Russell; Gunaydin, Hakan; Guzman-Perez, Angel; Huang, Hongbing; Huang, Liyue; Jarosh, Michael; Kornecook, Thomas; Kreiman, Charles R.; Ligutti, Joseph; La, Daniel S.; Lin, Min-Hwa Jasmine; Liu, Dong; Moyer, Bryan D.; Nguyen, Hanh N.; Peterson, Emily A.; Rose, Paul E.; Taborn, Kristin; Youngblood, Beth D.; Yu, Violeta; Fremeau, Robert T.Journal of Medicinal Chemistry (2017), 60 (14), 5969-5989CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Several reports have recently emerged regarding the identification of heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. The optimization of a series of internal NaV1.7 leads that address a no. of metabolic liabilities including bioactivation, PXR activation, as well as CYP3A4 induction and inhibition led to the identification of potent and selective inhibitors that demonstrated favorable pharmacokinetic profiles and were devoid of the aforementioned liabilities. The key to achieving this within a series prone to transporter-mediated clearance was the identification of a small range of optimal cLogD values and the discovery of subtle PXR SAR that was not lipophilicity dependent. This enabled the identification of compd. 20, which was advanced into a target engagement pharmacodynamic model where it exhibited robust reversal of histamine-induced scratching bouts in mice.
- 35Dittmar, M.; Lee, J. S.; Whig, K.; Segrist, E.; Li, M.; Kamalia, B.; Castellana, L.; Ayyanathan, K.; Cardenas-Diaz, F. L.; Morrisey, E. E.; Truitt, R.; Yang, W.; Jurado, K.; Samby, K.; Ramage, H.; Schultz, D. C.; Cherry, S. Drug repurposing screens reveal cell-type-specific entry pathways and FDA-approved drugs active against SARS-Cov-2. Cell Rep. 2021, 35 (1), 108959, DOI: 10.1016/j.celrep.2021.108959Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnsVWju7w%253D&md5=52eaa65b358385380f995e46d3c7703dDrug repurposing screens reveal cell-type-specific entry pathways and FDA-approved drugs active against SARS-Cov-2Dittmar, Mark; Lee, Jae Seung; Whig, Kanupriya; Segrist, Elisha; Li, Minghua; Kamalia, Brinda; Castellana, Lauren; Ayyanathan, Kasirajan; Cardenas-Diaz, Fabian L.; Morrisey, Edward E.; Truitt, Rachel; Yang, Wenli; Jurado, Kellie; Samby, Kirandeep; Ramage, Holly; Schultz, David C.; Cherry, SaraCell Reports (2021), 35 (1), 108959CODEN: CREED8; ISSN:2211-1247. (Cell Press)There is an urgent need for antivirals to treat the newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To identify new candidates, we screen a repurposing library of ~ 3,000 drugs. Screening in Vero cells finds few antivirals, while screening in human Huh7.5 cells validates 23 diverse antiviral drugs. Extending our studies to lung epithelial cells, we find that there are major differences in drug sensitivity and entry pathways used by SARS-CoV-2 in these cells. Entry in lung epithelial Calu-3 cells is pH independent and requires TMPRSS2, while entry in Vero and Huh7.5 cells requires low pH and triggering by acid-dependent endosomal proteases. Moreover, we find nine drugs are antiviral in respiratory cells, seven of which have been used in humans, and three are US Food and Drug Administration (FDA) approved, including cyclosporine. We find that the antiviral activity of cyclosporine is targeting Cyclophilin rather than calcineurin, revealing essential host targets that have the potential for rapid clin. implementation.
- 36Jang, W. D.; Jeon, S.; Kim, S.; Lee, S. Y. Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assay. Proc. Natl. Acad. Sci. U. S. A. 2021, 118 (30), e2024302118 DOI: 10.1073/pnas.2024302118Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1KmsbjN&md5=d37de60099bc07ebf242e4a17132e153Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assayJang, Woo Dae; Jeon, Sangeun; Kim, Seungtaek; Lee, Sang YupProceedings of the National Academy of Sciences of the United States of America (2021), 118 (30), e2024302118CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The COVID-19 pandemic caused by SARS-CoV-2 is an unprecedentedly significant health threat, prompting the need for rapidly developing antiviral drugs for the treatment. Drug repurposing is currently 1 of the most tangible options for rapidly developing drugs for emerging and reemerging viruses. In general, drug repurposing starts with virtual screening of approved drugs employing various computational methods. However, the actual hit rate of virtual screening is very low, and most of the predicted compds. are false positives. Here, we developed a strategy for virtual screening with much reduced false positives through incorporating predocking filtering based on shape similarity and postdocking filtering based on interaction similarity. We applied this advanced virtual screening approach to repurpose 6218 approved and clin. trial drugs for COVID-19. All 6218 compds. were screened against main protease and RNA-dependent RNA polymerase of SARS-CoV-2, resulting in 15 and 23 potential repurposed drugs, resp. Among them, 7 compds. can inhibit SARS-CoV-2 replication in Vero cells. Three of these drugs, emodin, omipalisib, and tipifarnib, show anti-SARS-CoV-2 activities in human lung cells, Calu-3. Notably, the activity of omipalisib is 200-fold higher than that of remdesivir in Calu-3. Furthermore, 3 drug combinations, omipalisib/remdesivir, tipifarnib/omipalisib, and tipifarnib/remdesivir, show strong synergistic effects in inhibiting SARS-CoV-2. Such drug combination therapy improves antiviral efficacy in SARS-CoV-2 infection and reduces the risk of each drug's toxicity. The drug repurposing strategy reported here will be useful for rapidly developing drugs for treating COVID-19 and other viruses.
- 37Choi, S. W.; Shin, J. S.; Park, S. J.; Jung, E.; Park, Y. G.; Lee, J.; Kim, S. J.; Park, H. J.; Lee, J. H.; Park, S. M.; Moon, S. H.; Ban, K.; Go, Y. Y. Antiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytes. Antiviral Res. 2020, 184, 104955, DOI: 10.1016/j.antiviral.2020.104955Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitl2nsbfJ&md5=2a4723ae20790ada7df5ecabe730a2bdAntiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytesChoi, Seong Woo; Shin, Jin Soo; Park, Soon-Jung; Jung, Eunhye; Park, Yun-Gwi; Lee, Jiho; Kim, Sung Joon; Park, Hun-Jun; Lee, Jung-Hoon; Park, Sung-Min; Moon, Sung-Hwan; Ban, Kiwon; Go, Yun YoungAntiviral Research (2020), 184 (), 104955CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is considered as the most significant global public health crisis of the century. Several drug candidates have been suggested as potential therapeutic options for COVID-19, including remdesivir, currently the only authorized drug for use under an Emergency Use Authorization. However, there is only limited information regarding the safety profiles of the proposed drugs, in particular drug-induced cardiotoxicity. Here, we evaluated the antiviral activity and cardiotoxicity of remdesivir using cardiomyocytes-derived from human pluripotent stem cells (hPSC-CMs) as an alternative source of human primary cardiomyocytes (CMs). In this study, remdesivir exhibited up to 60-fold higher antiviral activity in hPSC-CMs compared to Vero E6 cells; however, it also induced moderate cardiotoxicity in these cells. To gain further insight into the drug-induced arrhythmogenic risk, we assessed QT interval prolongation and automaticity of remdesivir-treated hPSC-CMs using a multielectrode array (MEA). As a result, the data indicated a potential risk of QT prolongation when remdesivir is used at concns. higher than the estd. peak plasma concn. Therefore, we conclude that close monitoring of the electrocardiog./QT interval should be advised in SARS-CoV-2-infected patients under remdesivir medication, in particular individuals with pre-existing heart conditions.
- 38Tiwari, S. K.; Wang, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Revealing Tissue-Specific SARS-CoV-2 Infection and Host Responses using Human Stem Cell-Derived Lung and Cerebral Organoids. Stem Cell Rep. 2021, 16 (3), 437– 445, DOI: 10.1016/j.stemcr.2021.02.005Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXls1ersbw%253D&md5=e12410b74f51084672e12fab3eb5230eRevealing tissue-specific SARS-CoV-2 infection and host responses using human stem cell-derived lung and cerebral organoidsTiwari, Shashi Kant; Wang, Shaobo; Smith, Davey; Carlin, Aaron F.; Rana, Tariq M.Stem Cell Reports (2021), 16 (3), 437-445CODEN: SCRTFL; ISSN:2213-6711. (Cell Press)COVID-19 is a transmissible respiratory disease caused by a novel coronavirus, SARS-CoV-2, and has become a global health emergency. There is an urgent need for robust and practical in vitro model systems to investigate viral pathogenesis. Here, we generated human induced pluripotent stem cell (iPSC)-derived lung organoids (LORGs), cerebral organoids (CORGs), neural progenitor cells (NPCs), neurons, and astrocytes. LORGs contg. epithelial cells, alveolar types 1 and 2, highly express ACE2 and TMPRSS2 and are permissive to SARS-CoV-2 infection. SARS-CoV-2 infection induces interferons, cytokines, and chemokines and activates crit. inflammasome pathway genes. Spike protein inhibitor, EK1 peptide, and TMPRSS2 inhibitors (camostat/nafamostat) block viral entry in LORGs. Conversely, CORGs, NPCs, astrocytes, and neurons express low levels of ACE2 and TMPRSS2 and correspondingly are not highly permissive to SARS-CoV-2 infection. Infection in neuronal cells activates TLR3/7, OAS2, complement system, and apoptotic genes. These findings will aid in understanding COVID-19 pathogenesis and facilitate drug discovery.
- 39Li, N.; Hui, H.; Bray, B.; Gonzalez, G. M.; Zeller, M.; Anderson, K. G.; Knight, R.; Smith, D.; Wang, Y.; Carlin, A. F.; Rana, T. M. METTL3 regulates viral m6A RNA modification and host cell innate immune responses during SARS-CoV-2 infection. Cell Rep. 2021, 35 (6), 109091, DOI: 10.1016/j.celrep.2021.109091Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVShu7zP&md5=938ee9a0e2de397723df26374d4e2612METTL3 regulates viral m6A RNA modification and host cell innate immune responses during SARS-CoV-2 infectionLi, Na; Hui, Hui; Bray, Bill; Gonzalez, Gwendolyn Michelle; Zeller, Mark; Anderson, Kristian G.; Knight, Rob; Smith, Davey; Wang, Yinsheng; Carlin, Aaron F.; Rana, Tariq M.Cell Reports (2021), 35 (6), 109091CODEN: CREED8; ISSN:2211-1247. (Cell Press)It is urgent and important to understand the relationship of the widespread severe acute respiratory syndrome coronavirus clade 2 (SARS-CoV-2) with host immune response and study the underlining mol. mechanism. N6-methylation of adenosine (m6A) in RNA regulates many physiol. and disease processes. Here, we investigate m6A modification of the SARS-CoV-2 gene in regulating the host cell innate immune response. Our data show that the SARS-CoV-2 virus has m6A modifications that are enriched in the 3' end of the viral genome. We find that depletion of the host cell m6A methyltransferase METTL3 decreases m6A levels in SARS-CoV-2 and host genes, and m6A redn. in viral RNA increases RIG-I binding and subsequently enhances the downstream innate immune signaling pathway and inflammatory gene expression. METTL3 expression is reduced and inflammatory genes are induced in patients with severe coronavirus disease 2019 (COVID-19). These findings will aid in the understanding of COVID-19 pathogenesis and the design of future studies regulating innate immunity for COVID-19 treatment.
- 40Wang, S.; Li, W.; Hui, H.; Tiwari, S. K.; Zhang, Q.; Croker, B. A.; Rawlings, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Cholesterol 25-Hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterol. EMBO J. 2020, 39 (21), e106057 DOI: 10.15252/embj.2020106057Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFyisbvL&md5=7b37216366d03b329695f0703c5e96dfCholesterol 25-Hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterolWang, Shaobo; Li, Wanyu; Hui, Hui; Tiwari, Shashi Kant; Zhang, Qiong; Croker, Ben A.; Rawlings, Stephen; Smith, Davey; Carlin, Aaron F.; Rana, Tariq M.EMBO Journal (2020), 39 (21), e106057CODEN: EMJODG; ISSN:0261-4189. (Wiley-VCH Verlag GmbH & Co. KGaA)Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2 and has spread across the globe. SARS-CoV-2 is a highly infectious virus with no vaccine or antiviral therapy available to control the pandemic; therefore, it is crucial to understand the mechanisms of viral pathogenesis and the host immune responses to SARS-CoV-2. SARS-CoV-2 is a new member of the betacoronavirus genus like other closely related viruses including SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Both SARS-CoV and MERS-CoV have caused serious outbreaks and epidemics in the past 18 yr. We report that 1 of the interferon-stimulated genes (ISGs), cholesterol 25-hydroxylase (CH25H), is induced by SARS-CoV-2 infection in vitro and in COVID-19-infected patients. CH25H converts cholesterol to 25-hydroxycholesterol (25HC) and 25HC shows broad anti-coronavirus activity by blocking membrane fusion. Furthermore, 25HC inhibits USA-WA1/2020 SARS-CoV-2 infection in lung epithelial cells and viral entry in human lung organoids. Mechanistically, 25HC inhibits viral membrane fusion by activating the ER-localized acyl-CoA:cholesterol acyltransferase (ACAT) which leads to the depletion of accessible cholesterol from the plasma membrane. Altogether, our results shed light on a potentially broad antiviral mechanism by 25HC through depleting accessible cholesterol on the plasma membrane to suppress virus-cell fusion. Since 25HC is a natural product with no known toxicity at effective concns., it provides a potential therapeutic candidate for COVID-19 and emerging viral diseases in the future.
- 41Ma, C.; Hu, Y.; Townsend, J. A.; Lagarias, P. I.; Marty, M. T.; Kolocouris, A.; Wang, J. Ebselen, Disulfiram, Carmofur, PX-12, Tideglusib, and Shikonin Are Nonspecific Promiscuous SARS-CoV-2 Main Protease Inhibitors. ACS Pharmacol Transl Sci. 2020, 3 (6), 1265– 1277, DOI: 10.1021/acsptsci.0c00130Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVajsrjK&md5=29d0e69db20f681ed4ef7cddd153cc6dEbselen, Disulfiram, Carmofur, PX-12, Tideglusib, and Shikonin Are Nonspecific Promiscuous SARS-CoV-2 Main Protease InhibitorsMa, Chunlong; Hu, Yanmei; Townsend, Julia Alma; Lagarias, Panagiotis I.; Marty, Michael Thomas; Kolocouris, Antonios; Wang, JunACS Pharmacology & Translational Science (2020), 3 (6), 1265-1277CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)Among the drug targets being investigated for SARS-CoV-2, the viral main protease (Mpro) is one of the most extensively studied. Mpro is a cysteine protease that hydrolyzes the viral polyprotein at more than 11 sites. It is highly conserved and has a unique substrate preference for glutamine in the P1 position. Therefore, Mpro inhibitors are expected to have broad-spectrum antiviral activity and a high selectivity index. Structurally diverse compds. have been reported as Mpro inhibitors. In this study, we investigated the mechanism of action of six previously reported Mpro inhibitors, ebselen, disulfiram, tideglusib, carmofur, shikonin, and PX-12, using a consortium of techniques including FRET-based enzymic assay, thermal shift assay, native mass spectrometry, cellular antiviral assays, and mol. dynamics simulations. Collectively, the results showed that the inhibition of Mpro by these six compds. is nonspecific and that the inhibition is abolished or greatly reduced with the addn. of reducing reagent 1,4-dithiothreitol (DTT). Without DTT, these six compds. inhibit not only Mpro but also a panel of viral cysteine proteases including SARS-CoV-2 papain-like protease and 2Apro and 3Cpro from enterovirus A71 (EV-A71) and EV-D68. However, none of the compds. inhibits the viral replication of EV-A71 or EV-D68, suggesting that the enzymic inhibition potency IC50 values obtained in the absence of DTT cannot be used to faithfully predict their cellular antiviral activity. Overall, we provide compelling evidence suggesting that these six compds. are nonspecific SARS-CoV-2 Mpro inhibitors and urge the scientific community to be stringent with hit validation.
- 42Weglarz-Tomczak, E.; Tomczak, J. M.; Talma, M.; Burda-Grabowska, M.; Giurg, M.; Brul, S. Identification of ebselen and its analogues as potent covalent inhibitors of papain-like protease from SARS-CoV-2. Sci. Rep. 2021, 11 (1), 3640, DOI: 10.1038/s41598-021-83229-6Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktVOntbY%253D&md5=bb905ef94cebd529c4fc53d196adc5a4Identification of ebselen and its analogues as potent covalent inhibitors of papain-like protease from SARS-CoV-2Weglarz-Tomczak, Ewelina; Tomczak, Jakub M.; Talma, Michal; Burda-Grabowska, Malgorzata; Giurg, Miroslaw; Brul, StanleyScientific Reports (2021), 11 (1), 3640CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)An efficient treatment against a COVID-19 disease, caused by the novel coronavirus SARS-CoV-2 (CoV2), remains a challenge. The papain-like protease (PLpro) from the human coronavirus is a protease that plays a crit. role in virus replication. Moreover, CoV2 uses this enzyme to modulate the host's immune system to its own benefit. Therefore, it represents a highly promising target for the development of antiviral drugs. We used Approx. Bayesian Computation tools, mol. modeling and enzyme activity studies to identify highly active inhibitors of the PLpro. We discovered organoselenium compds., ebselen and its structural analogs, as a novel approach for inhibiting the activity of PLproCoV2. Furthermore, we identified, for the first time, inhibitors of PLproCoV2 showing potency in the nanomolar range. Moreover, we found a difference between PLpro from SARS and CoV2 that can be correlated with the diverse dynamics of their replication, and, putatively to disease progression.
- 43Freitas, B. T.; Durie, I. A.; Murray, J.; Longo, J. E.; Miller, H. C.; Crich, D.; Hogan, R. J.; Tripp, R. A.; Pegan, S. D. Characterization and Noncovalent Inhibition of the Deubiquitinase and deISGylase Activity of SARS-CoV-2 Papain-Like Protease. ACS Infect. Dis. 2020, 6 (8), 2099– 2109, DOI: 10.1021/acsinfecdis.0c00168Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpslylsbs%253D&md5=314ea59a92d60345f982e78e31e5b569Characterization and Noncovalent Inhibition of the Deubiquitinase and deISGylase Activity of SARS-CoV-2 Papain-Like ProteaseFreitas, Brendan T.; Durie, Ian A.; Murray, Jackelyn; Longo, Jaron E.; Miller, Holden C.; Crich, David; Hogan, Robert Jeff; Tripp, Ralph A.; Pegan, Scott D.ACS Infectious Diseases (2020), 6 (8), 2099-2109CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent for COVID-19, is a novel human betacoronavirus that is rapidly spreading worldwide. The outbreak currently includes over 3.7 million cases and 260,000 fatalities. As a betacoronavirus, SARS-CoV-2 encodes for a papain-like protease (PLpro) that is likely responsible for cleavage of the coronavirus (CoV) viral polypeptide. The PLpro is also responsible for suppression of host innate immune responses by virtue of its ability to reverse host ubiquitination and ISGylation events. Here, the biochem. activity of SARS-CoV-2 PLpro against ubiquitin (Ub) and interferon-stimulated gene product 15 (ISG15) substrates is evaluated, revealing that the protease has a marked redn. in its ability to process K48 linked Ub substrates compared to its counterpart in SARS-CoV. Addnl., its substrate activity more closely mirrors that of the PLpro from the Middle East respiratory syndrome coronavirus and prefers ISG15s from certain species including humans. Addnl., naphthalene based PLpro inhibitors are shown to be effective at halting SARS-CoV-2 PLpro activity as well as SARS-CoV-2 replication.
- 44Shin, D.; Mukherjee, R.; Grewe, D.; Bojkova, D.; Baek, K.; Bhattacharya, A.; Schulz, L.; Widera, M.; Mehdipour, A. R.; Tascher, G.; Geurink, P. P.; Wilhelm, A.; van der Heden van Noort, G. J.; Ovaa, H.; Muller, S.; Knobeloch, K. P.; Rajalingam, K.; Schulman, B. A.; Cinatl, J.; Hummer, G.; Ciesek, S.; Dikic, I. Papain-like protease regulates SARS-CoV-2 viral spread and innate immunity. Nature 2020, 587 (7835), 657– 662, DOI: 10.1038/s41586-020-2601-5Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVSqu7nP&md5=d6e55cd2e91c4a55f19abf2652973aabPapain-like protease regulates SARS-CoV-2 viral spread and innate immunityShin, Donghyuk; Mukherjee, Rukmini; Grewe, Diana; Bojkova, Denisa; Baek, Kheewoong; Bhattacharya, Anshu; Schulz, Laura; Widera, Marek; Mehdipour, Ahmad Reza; Tascher, Georg; Geurink, Paul P.; Wilhelm, Alexander; van der Heden van Noort, Gerbrand J.; Ovaa, Huib; Mueller, Stefan; Knobeloch, Klaus-Peter; Rajalingam, Krishnaraj; Schulman, Brenda A.; Cinatl, Jindrich; Hummer, Gerhard; Ciesek, Sandra; Dikic, IvanNature (London, United Kingdom) (2020), 587 (7835), 657-662CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The papain-like protease PLpro is an essential coronavirus enzyme that is required for processing viral polyproteins to generate a functional replicase complex and enable viral spread. PLpro is also implicated in cleaving proteinaceous post-translational modifications on host proteins as an evasion mechanism against host antiviral immune responses. We perform biochem., structural, and functional characterization of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PLpro (SCoV2-PLpro) and outline differences with SARS-CoV PLpro (SCoV-PLpro) in regulation of host interferon and NF-κB pathways. SCoV2-PLpro and SCoV-PLpro share 83% sequence identity but exhibit different host substrate preferences; SCoV2-PLpro preferentially cleaves the ubiquitin-like interferon-stimulated gene 15 protein (ISG15), whereas SCoV-PLpro predominantly targets ubiquitin chains. The crystal structure of SCoV2-PLpro in complex with ISG15 reveals distinctive interactions with the N-terminal ubiquitin-like domain of ISG15, highlighting the high affinity and specificity of these interactions. Furthermore, upon infection, SCoV2-PLpro contributes to the cleavage of ISG15 from interferon responsive factor 3 (IRF3) and attenuates type I interferon responses. Notably, inhibition of SCoV2-PLpro with GRL-0617 impairs the virus-induced cytopathogenic effect, maintains the antiviral interferon pathway and reduces viral replication in infected cells. These results highlight a potential dual therapeutic strategy in which targeting of SCoV2-PLpro can suppress SARS-CoV-2 infection and promote antiviral immunity.
- 45Klemm, T.; Ebert, G.; Calleja, D. J; Allison, C. C; Richardson, L. W; Bernardini, J. P; Lu, B. G.; Kuchel, N. W; Grohmann, C.; Shibata, Y.; Gan, Z. Y.; Cooney, J. P; Doerflinger, M.; Au, A. E; Blackmore, T. R; Heden van Noort, G. J; Geurink, P. P; Ovaa, H.; Newman, J.; Riboldi-Tunnicliffe, A.; Czabotar, P. E; Mitchell, J. P; Feltham, R.; Lechtenberg, B. C; Lowes, K. N; Dewson, G.; Pellegrini, M.; Lessene, G.; Komander, D. Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2. EMBO J. 2020, 39 (18), e106275 DOI: 10.15252/embj.2020106275Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1KgsrbE&md5=3d6a13fc99f05c4fdd6666e8fa72bbd7Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2Klemm, Theresa; Ebert, Gregor; Calleja, Dale J.; Allison, Cody C.; Richardson, Lachlan W.; Bernardini, Jonathan P.; Lu, Bernadine G. C.; Kuchel, Nathan W.; Grohmann, Christoph; Shibata, Yuri; Gan, Zhong Yan; Cooney, James P.; Doerflinger, Marcel; Au, Amanda E.; Blackmore, Timothy R.; van der Heden van Noort, Gerbrand J.; Geurink, Paul P.; Ovaa, Huib; Newman, Janet; Riboldi-Tunnicliffe, Alan; Czabotar, Peter E.; Mitchell, Jeffrey P.; Feltham, Rebecca; Lechtenberg, Bernhard C.; Lowes, Kym N.; Dewson, Grant; Pellegrini, Marc; Lessene, Guillaume; Komander, DavidEMBO Journal (2020), 39 (18), e106275CODEN: EMJODG; ISSN:0261-4189. (Wiley-VCH Verlag GmbH & Co. KGaA)The SARS-CoV-2 coronavirus encodes an essential papain-like protease domain as part of its non-structural protein (nsp)-3, namely SARS2 PLpro, that cleaves the viral polyprotein, but also removes ubiquitin-like ISG15 protein modifications as well as, with lower activity, Lys48-linked polyubiquitin. Structures of PLpro bound to ubiquitin and ISG15 reveal that the S1 ubiquitin-binding site is responsible for high ISG15 activity, while the S2 binding site provides Lys48 chain specificity and cleavage efficiency. To identify PLpro inhibitors in a repurposing approach, screening of 3,727 unique approved drugs and clin. compds. against SARS2 PLpro identified no compds. that inhibited PLpro consistently or that could be validated in counterscreens. More promisingly, non-covalent small mol. SARS PLpro inhibitors also target SARS2 PLpro, prevent self-processing of nsp3 in cells and display high potency and excellent antiviral activity in a SARS-CoV-2 infection model.
- 46Liu, G.; Lee, J.-H.; Parker, Z. M.; Acharya, D.; Chiang, J. J.; van Gent, M.; Riedl, W.; Davis-Gardner, M. E.; Wies, E.; Chiang, C.; Gack, M. U. ISG15-dependent activation of the sensor MDA5 is antagonized by the SARS-CoV-2 papain-like protease to evade host innate immunity. Nature Microbiology 2021, 6, 467– 478, DOI: 10.1038/s41564-021-00884-1Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXms1Onsbs%253D&md5=a74cf2f655bfac4c2e8516b0f9f0f4e0ISG15-dependent activation of the sensor MDA5 is antagonized by the SARS-CoV-2 papain-like protease to evade host innate immunityLiu, GuanQun; Lee, Jung-Hyun; Parker, Zachary M.; Acharya, Dhiraj; Chiang, Jessica J.; van Gent, Michiel; Riedl, William; Davis-Gardner, Meredith E.; Wies, Effi; Chiang, Cindy; Gack, Michaela U.Nature Microbiology (2021), 6 (4), 467-478CODEN: NMAICH; ISSN:2058-5276. (Nature Research)Abstr.: Activation of the RIG-I-like receptors, retinoic-acid inducible gene I (RIG-I) and melanoma differentiation-assocd. protein 5 (MDA5), establishes an antiviral state by upregulating interferon (IFN)-stimulated genes (ISGs). Among these is ISG15, the mechanistic roles of which in innate immunity still remain enigmatic. In the present study, we report that ISG15 conjugation is essential for antiviral IFN responses mediated by the viral RNA sensor MDA5. ISGylation of the caspase activation and recruitment domains of MDA5 promotes its oligomerization and thereby triggers activation of innate immunity against a range of viruses, including coronaviruses, flaviviruses and picornaviruses. The ISG15-dependent activation of MDA5 is antagonized through direct de-ISGylation mediated by the papain-like protease of SARS-CoV-2, a recently emerged coronavirus that has caused the COVID-19 pandemic. Our work demonstrates a crucial role for ISG15 in the MDA5-mediated antiviral response, and also identifies a key immune evasion mechanism of SARS-CoV-2, which may be targeted for the development of new antivirals and vaccines to combat COVID-19.
- 47Jacobson, M. P.; Friesner, R. A.; Xiang, Z.; Honig, B. On the role of the crystal environment in determining protein side-chain conformations. J. Mol. Biol. 2002, 320 (3), 597– 608, DOI: 10.1016/S0022-2836(02)00470-9Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XltVKmu70%253D&md5=006de6bd2d0f233ab32d6798dc1a3fbcOn the Role of the Crystal Environment in Determining Protein Side-chain ConformationsJacobson, Matthew P.; Friesner, Richard A.; Xiang, Zhexin; Honig, BarryJournal of Molecular Biology (2002), 320 (3), 597-608CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Science Ltd.)The role of crystal packing in detg. the obsd. conformations of amino acid side-chains in protein crystals is investigated by (1) anal. of a database of proteins that have been crystd. in different unit cells (space group or unit cell dimensions) and (2) theor. predictions of side-chain conformations with the crystal environment explicitly represented. Both of these approaches indicate that the crystal environment plays an important role in detg. the conformations of polar side-chains on the surfaces of proteins. Inclusion of the crystal environment permits a more sensitive measurement of the achievable accuracy of side-chain prediction programs, when validating against structures obtained by x-ray crystallog. Our side-chain prediction program uses an all-atom force field and a Generalized Born model of solvation and is thus capable of modeling simple packing effects (i.e. van der Waals interactions), electrostatic effects, and desolvation, which are all important mechanisms by which the crystal environment impacts obsd. side-chain conformations. Our results are also relevant to the understanding of changes in side-chain conformation that may result from ligand docking and protein-protein assocn., insofar as the results reveal how side-chain conformations change in response to their local environment.
- 48Jacobson, M. P.; Pincus, D. L.; Rapp, C. S.; Day, T. J.; Honig, B.; Shaw, D. E.; Friesner, R. A. A hierarchical approach to all-atom protein loop prediction. Proteins: Struct., Funct., Genet. 2004, 55 (2), 351– 367, DOI: 10.1002/prot.10613Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjtFKhsrc%253D&md5=e0eff655eeefb30ea00ae041ea9099c8A hierarchical approach to all-atom protein loop predictionJacobson, Matthew P.; Pincus, David L.; Rapp, Chaya S.; Day, Tyler J. F.; Honig, Barry; Shaw, David E.; Friesner, Richard A.Proteins: Structure, Function, and Bioinformatics (2004), 55 (2), 351-367CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The application of all-atom force fields (and explicit or implicit solvent models) to protein homol.-modeling tasks such as side-chain and loop prediction remains challenging both because of the expense of the individual energy calcns. and because of the difficulty of sampling the rugged all-atom energy surface. Here the authors address this challenge for the problem of loop prediction through the development of numerous new algorithms, with an emphasis on multiscale and hierarchical techniques. As a first step in evaluating the performance of the authors' loop prediction algorithm, the authors have applied it to the problem of reconstructing loops in native structures; the authors also explicitly include crystal packing to provide a fair comparison with crystal structures. In brief, large nos. of loops are generated by using a dihedral angle-based buildup procedure followed by iterative cycles of clustering, side-chain optimization, and complete energy minimization of selected loop structures. The authors evaluate this method by the largest test set yet used for validation of a loop prediction method, with a total of 833 loops ranging from 4 to 12 residues in length. Av./median backbone root-mean-square deviations (RMSDs) to the native structures (superimposing the body of the protein, not the loop itself) are 0.42/0.24 Å for 5 residue loops, 1.00/0.44 Å for 8 residue loops, and 2.47/1.83 Å for 11 residue loops. Median RMSDs are substantially lower than the avs. because of a small no. of outliers; the causes of these failures are examd. in some detail, and many can be attributed to errors in assignment of protonation states of titratable residues, omission of ligands from the simulation, and, in a few cases, probable errors in the exptl. detd. structures. When these obvious problems in the data sets are filtered out, av. RMSDs to the native structures improve to 0.43 Å for 5 residue loops, 0.84 Å for 8 residue loops, and 1.63 Å for 11 residue loops. In the vast majority of cases, the method locates energy min. that are lower than or equal to that of the minimized native loop, thus indicating that sampling rarely limits prediction accuracy. The overall results are, to the authors' knowledge, the best reported to date, and the authors attribute this success to the combination of an accurate all-atom energy function, efficient methods for loop buildup and side-chain optimization, and, esp. for the longer loops, the hierarchical refinement protocol.
- 49Donovan, S. F.; Pescatore, M. C. Method for measuring the logarithm of the octanol-water partition coefficient by using short octadecyl-poly(vinyl alcohol) high-performance liquid chromatography columns. J. Chromatogr A 2002, 952 (1–2), 47– 61, DOI: 10.1016/S0021-9673(02)00064-XGoogle Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XisV2gs7s%253D&md5=4c4aa02a23c1a496d3a9107d43329629Method for measuring the logarithm of the octanol-water partition coefficient by using short octadecyl-poly(vinyl alcohol) high-performance liquid chromatography columnsDonovan, Stephen F.; Pescatore, Mark C.Journal of Chromatography A (2002), 952 (1-2), 47-61CODEN: JCRAEY; ISSN:0021-9673. (Elsevier Science B.V.)A simple, quick, versatile and inexpensive HPLC method to est. the logarithm of the octanol-water partition coeff. (log Pow) employing a methanol-water gradient and a short octadecyl-poly(vinyl alc.) (ODP) column is described. This method is different from published HPLC-based log Pow methods because it uses retention times from a rapid methanol-water gradient to directly generate log Pow ests., rather than from a series of isocratic mixts. extrapolated to 100% water. These HPLC log Pow values have good precision and correlate well with traditional shake-flask log Pow values. If necessary, the log Pow detn. (including replications) can easily be carried out using only a milligram of sample. By suppressing ionization of acids and bases by the use of a buffer in the aq. phase, the method can measure the log Pow of neutral org. mols. at any pH between 2 and 13. The method can be used with impure material and is rapid, 7 min per run and 4 min equilibration; it lends itself to and has been utilized for high-throughput hydrophobicity detns. (we have now carried out thousands of HPLC log Pow measurements by this method).
- 50Box, K.; Bevan, C.; Comer, J.; Hill, A.; Allen, R.; Reynolds, D. High-throughput measurement of pKa values in a mixed-buffer linear pH gradient system. Anal. Chem. 2003, 75 (4), 883– 892, DOI: 10.1021/ac020329yGoogle Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjt1yksg%253D%253D&md5=4050d66a7c92550c1efd346e76c392d9High-Throughput Measurement of pKa Values in a Mixed-Buffer Linear pH Gradient SystemBox, Karl; Bevan, Christopher; Comer, John; Hill, Alan; Allen, Ruth; Reynolds, DerekAnalytical Chemistry (2003), 75 (4), 883-892CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A procedure is described for measuring pKa values in a short time, e.g., 4 min/assay. Samples, as 10 mM solns., are prepd. in DMSO in 96-well plates. A flowing pH gradient is produced by mixing two buffer solns. contg. mixts. of weak acids and bases that do not absorb significantly in the UV above 250 nm. The sample soln. is dild. with water and then injected directly into the flowing gradient, which then passes through a diode array spectrophotometer measuring in the UV wavelength range. The buffer has been formulated so that its acid-base titrn. curve is linear over a wide pH range, such that the pH of the gradient is a linear function of time. The soln. pH in the measurement flow cell is therefore proportional to the time elapsed since the start of gradient generation. The sample's pKa values are calcd. from the change in UV absorbance at multiple wavelengths as a function of pH. The pKa values of 71 drugs have been measured, and results compare well with values measured by pH-metric or traditional UV methods. Rules are suggested for the rapid inspection of data and the choice of method for the calcn. of pKa from the data.
Cited By
This article is cited by 74 publications.
- Zhihao Gu, Yong Yan, Hanwen Liu, Di Wu, Hequan Yao, Kejiang Lin, Xuanyi Li. Discovery of Covalent Lead Compounds Targeting 3CL Protease with a Lateral Interactions Spiking Neural Network. Journal of Chemical Information and Modeling 2024, 64
(8)
, 3047-3058. https://doi.org/10.1021/acs.jcim.3c01900
- Katarzyna Macegoniuk, Wojciech Tabor, Luca Mazzei, Michele Cianci, Mirosław Giurg, Kamila Olech, Małgorzata Burda-Grabowska, Rafał Kaleta, Agnieszka Grabowiecka, Artur Mucha, Stefano Ciurli, Łukasz Berlicki. Optimized Ebselen-Based Inhibitors of Bacterial Ureases with Nontypical Mode of Action. Journal of Medicinal Chemistry 2023, 66
(3)
, 2054-2063. https://doi.org/10.1021/acs.jmedchem.2c01799
- Xi Feng, Lixia Pan, Zhiyu Qian, Dongyu Liu, Xin Guan, Li Feng, Bin Song, Xi Xu, Ninghua Tan, Yi Ma, Zhiyu Li, Zhe Wang, Jinlei Bian. Discovery of Selenium-Containing STING Agonists as Orally Available Antitumor Agents. Journal of Medicinal Chemistry 2022, 65
(22)
, 15048-15065. https://doi.org/10.1021/acs.jmedchem.2c00634
- Gabriele La Monica, Alessia Bono, Antonino Lauria, Annamaria Martorana. Targeting SARS-CoV-2 Main Protease for Treatment of COVID-19: Covalent Inhibitors Structure–Activity Relationship Insights and Evolution Perspectives. Journal of Medicinal Chemistry 2022, 65
(19)
, 12500-12534. https://doi.org/10.1021/acs.jmedchem.2c01005
- Xin-Xin Liang, Xing-Jie Zhang, Ying-Xin Zhao, Jian Feng, Jie-Chun Zeng, Qiang-Qiang Shi, Joseph Sakah Kaunda, Xiao-Li Li, Wei-Guang Wang, Wei-Lie Xiao. Aspulvins A–H, Aspulvinone Analogues with SARS-CoV-2 Mpro Inhibitory and Anti-inflammatory Activities from an Endophytic Cladosporium sp.. Journal of Natural Products 2022, 85
(4)
, 878-887. https://doi.org/10.1021/acs.jnatprod.1c01003
- Wei Hou, Hongtao Xu. Incorporating Selenium into Heterocycles and Natural Products─From Chemical Properties to Pharmacological Activities. Journal of Medicinal Chemistry 2022, 65
(6)
, 4436-4456. https://doi.org/10.1021/acs.jmedchem.1c01859
- Nouri Neamati. Advances Toward COVID-19 Therapies Special Issue. Journal of Medicinal Chemistry 2022, 65
(4)
, 2713-2715. https://doi.org/10.1021/acs.jmedchem.2c00178
- Abdelali Chihab, Nabil El Brahmi, Abdelmoula El Abbouchi, Abdelaziz El Alaoui, Mostapha Bousmina, Elmostafa El Fahime, Saïd El Kazzouli. Synthesis, characterization, proteolytic activity inhibition, ADMET prediction, and molecular docking studies of novel indole derivatives as potential SARS-CoV-2 protease inhibitors. Journal of Molecular Structure 2025, 1323 , 140707. https://doi.org/10.1016/j.molstruc.2024.140707
- Aditya Trivedi, Tushar Kushwaha, Ishani, Sudhanshu Vrati, Dharmender Gupta, Sarala Rani Kayampeta, Mohammad Khalid Parvez, Krishna Kishore Inampudi, Mohan Babu Appaiahgari, Deepak Sehgal. Psoralidin acts as a dual protease inhibitor against
PL
pro
and M
pro
of
SARS
‐
CoV
‐2. The FEBS Journal 2025, 67 https://doi.org/10.1111/febs.17380
- Nabeel Ahmed, Aimen Sajid, Taseer Ahmad, Muhammad Ajmal Shah, Abdul Jabbar Shah. Coumarins as potential inhibitors targeting SARS-CoV-2. 2025, 151-182. https://doi.org/10.1016/B978-0-443-18757-5.00006-2
- Ilma Shakeel, Aaliya Taiyab, Aanchal Rathi, Taj Mohammad, Md. Imtaiyaz Hassan. Insights into SARS-CoV-2 structure: Approaches and action of SARS-CoV-2 main protease inhibitor. 2025, 89-117. https://doi.org/10.1016/B978-0-443-19170-1.00037-1
- S. k. Erfanul Haque, Sukanta Bhadra, Nishith Kumar Pal. Exploring potential therapeutic candidates against COVID-19: a molecular docking study. Discover Molecules 2024, 1
(1)
https://doi.org/10.1007/s44345-024-00005-5
- Kristine Mae H. Bonalos, Khent Primo Alcantara, Pornchai Rojsitthisak, Ross D. Vasquez. Comprehensive In silico and In vitro evaluation of Plectranthus amboinicus essential oil: A promising inhibitor of SARS-CoV-2 B.1.1.529 Omicron variant, proinflammatory biomarkers and diabetes-related enzymes. Industrial Crops and Products 2024, 222 , 119905. https://doi.org/10.1016/j.indcrop.2024.119905
- Luca Sancineto, Francesca Mangiavacchi, Agnieszka Dabrowska, Agata J. Pacuła-Miszewska, Magdalena Obieziurska-Fabisiak, Cecilia Scimmi, Veronica Ceccucci, Juan Kong, Yao Zhao, Gianluca Ciancaleoni, Vanessa Nascimento, Bruno Rizzuti, Marco Bortoli, Laura Orian, Anna Kula-Pacurar, Haitao Yang, Jacek Ścianowski, Ying Lei, Krzysztof Pyrc, Claudio Santi. New insights in the mechanism of the SARS-CoV-2 Mpro inhibition by benzisoselenazolones and diselenides. Scientific Reports 2024, 14
(1)
https://doi.org/10.1038/s41598-024-75519-6
- Chunru Liu, Xiaoqing Dong, Pinghuang Liu, Xi Lin. Advances in porcine respiratory and intestinal organoids: status and potential application for virus infections. One Health Advances 2024, 2
(1)
https://doi.org/10.1186/s44280-024-00052-0
- Md. Kaderi Kibria, Md. Ahad Ali, Md. Nurul Haque Mollah, . Exploring bacterial key genes and therapeutic agents for breast cancer among the Ghanaian female population: Insights from In Silico analyses. PLOS ONE 2024, 19
(11)
, e0312493. https://doi.org/10.1371/journal.pone.0312493
- Mianling Yang, Myoung Kyu Lee, Shenghua Gao, Letian Song, Hye‐Yeon Jang, Inseong Jo, Chun‐Chiao Yang, Katharina Sylvester, Chunkyu Ko, Shuo Wang, Bing Ye, Kai Tang, Junyi Li, Manyu Gu, Christa E. Müller, Norbert Sträter, Xinyong Liu, Meehyein Kim, Peng Zhan. Miniaturized Modular Click Chemistry‐enabled Rapid Discovery of Unique SARS‐CoV‐2 M
pro
Inhibitors With Robust Potency and Drug‐like Profile. Advanced Science 2024, 11
(43)
https://doi.org/10.1002/advs.202404884
- Asma Fatima, Anupriya M. Geethakumari, Wesam S. Ahmed, Kabir H. Biswas. A potential allosteric inhibitor of SARS-CoV-2 main protease (Mpro) identified through metastable state analysis. Frontiers in Molecular Biosciences 2024, 11 https://doi.org/10.3389/fmolb.2024.1451280
- Borwornlak Toopradab, Wanting Xie, Lian Duan, Kowit Hengphasatporn, Ryuhei Harada, Silpsiri Sinsulpsiri, Yasuteru Shigeta, Liyi Shi, Phornphimon Maitarad, Thanyada Rungrotmongkol. Machine learning-based QSAR and LB-PaCS-MD guided design of SARS-CoV-2 main protease inhibitors. Bioorganic & Medicinal Chemistry Letters 2024, 110 , 129852. https://doi.org/10.1016/j.bmcl.2024.129852
- Ying Kuan, Hsu-Feng Chu, Pang-Hung Hsu, Kai-Cheng Hsu, Ta-Hsien Lin, Chun-Hsiang Huang, Wei-Yi Chen. Disulfiram inhibits coronaviral main protease by conjugating to its substrate entry site. International Journal of Biological Macromolecules 2024, 276 , 133955. https://doi.org/10.1016/j.ijbiomac.2024.133955
- Christos Papaneophytou. Breaking the Chain: Protease Inhibitors as Game Changers in Respiratory Viruses Management. International Journal of Molecular Sciences 2024, 25
(15)
, 8105. https://doi.org/10.3390/ijms25158105
- Lei Han, Bing Wang, Kunhui Sun, Muqadas Sitara, Meifang Li, Ping Wang, Ning Chen, Xie-an Yu, Jiangwei Tian. A SARS-CoV-2 M
pro
fluorescent sensor for exploring pharmacodynamic substances from traditional Chinese medicine. The Analyst 2024, 149
(13)
, 3585-3595. https://doi.org/10.1039/D4AN00372A
- Ya-Qi Xiao, Jiao Long, Shuang-Shuang Zhang, Yuan-Yuan Zhu, Shuang-Xi Gu. Non-peptidic inhibitors targeting SARS-CoV-2 main protease: A review. Bioorganic Chemistry 2024, 147 , 107380. https://doi.org/10.1016/j.bioorg.2024.107380
- Hezha O. Rasul, Noel Vinay Thomas, Dlzar D. Ghafour, Bakhtyar K. Aziz, Guillermo Salgado M., L. H. Mendoza-Huizar, Lorena Gerli Candia. Searching possible SARS-CoV-2 main protease inhibitors in constituents from herbal medicines using
in silico
studies. Journal of Biomolecular Structure and Dynamics 2024, 42
(8)
, 4234-4248. https://doi.org/10.1080/07391102.2023.2220040
- Vimal K. Jain, K. Indira Priyadarsini. Selenium compounds as promising antiviral agents. New Journal of Chemistry 2024, 48
(15)
, 6534-6552. https://doi.org/10.1039/D3NJ05180C
- Hongtao Li, Meng Sun, Fuzhi Lei, Jinfeng Liu, Xixiang Chen, Yaqi Li, Ying Wang, Jiani Lu, Danmei Yu, Yueqiu Gao, Jianrong Xu, Hongzhuan Chen, Man Li, Zhigang Yi, Xiao He, Lili Chen. Methyl rosmarinate is an allosteric inhibitor of SARS-CoV-2 3 CL protease as a potential candidate against SARS-cov-2 infection. Antiviral Research 2024, 224 , 105841. https://doi.org/10.1016/j.antiviral.2024.105841
- Md. Kaderi Kibria, Md. Ahad Ali, Muhammad Yaseen, Imran Ahmad Khan, Mashooq Ahmad Bhat, Md. Ariful Islam, Rashidul Alam Mahumud, Md. Nurul Haque Mollah. Discovery of Bacterial Key Genes from 16S rRNA-Seq Profiles That Are Associated with the Complications of SARS-CoV-2 Infections and Provide Therapeutic Indications. Pharmaceuticals 2024, 17
(4)
, 432. https://doi.org/10.3390/ph17040432
- Prabuddha Bhattacharya, Anirban Mandal. Identification of amentoflavone as a potent SARS-CoV-2 M
pro
inhibitor: a combination of computational studies and in vitro biological evaluation. Journal of Biomolecular Structure and Dynamics 2024, 266 , 1-19. https://doi.org/10.1080/07391102.2024.2304676
- Samuel J.M. Santos, Antoninho Valentini. In silico investigation of Komaroviquinone as a potential inhibitor of SARS-CoV-2 main protease (Mpro): Molecular docking, molecular dynamics, and QM/MM approaches. Journal of Molecular Graphics and Modelling 2024, 126 , 108662. https://doi.org/10.1016/j.jmgm.2023.108662
- Frank Eric Tatsing Foka, Hazel Tumelo Mufhandu, . Predictive Assessment of the Antiviral Properties of
Imperata cylindrica
against SARS‐CoV‐2. Advances in Virology 2024, 2024
(1)
https://doi.org/10.1155/2024/8598708
- Seshadri Reddy Varikasuvu, Subodh Kumar, Munikumar Manne, Mona Lisa, Saurabh Varshney, Pratima Gupta, Ashoo Grover, Vanita Lal, Harminder Singh, Shiv Kumar Mudgal, Konda Venkata Nagaraju. Covid-19 Clinical Outcomes and N-Acetylcysteine (Covinac Study): A Grade Compliant Meta-Analysis of Randomized Controlled Trials, Molecular Docking, and Dynamics Simulations with Mpro of Sars-Cov-2. 2024https://doi.org/10.2139/ssrn.4679821
- Xiaoli Shen, Hao Zhang, Pengyin Zhang, Xuerui Zhao, Chang Liu, Jianan Ju, Aijun Liu, Song Wang. The Inhibitory Mechanism of Sars-Cov-2 by Small Molecules of the Condensed Amino Thiourea Scaffold Based on Molecular Dynamics Simulation. 2024https://doi.org/10.2139/ssrn.4961580
- Kailasam Saravana Mani, Sundarraj Rajamanikandan, Balakrishnan Prabha. Discovery, synthesis and
in silico
approach of pyrrolo [3,4-
c
]pyrroles as SARS-CoV-2 m
pro
inhibitors. Journal of Biomolecular Structure and Dynamics 2023, 41
(22)
, 13454-13465. https://doi.org/10.1080/07391102.2023.2174599
- Ssemuyiga Charles, Mulumba Pius Edgar, Rajani Kanta Mahapatra. Artificial intelligence based virtual screening study for competitive and allosteric inhibitors of the SARS-CoV-2 main protease. Journal of Biomolecular Structure and Dynamics 2023, 41
(24)
, 15286-15304. https://doi.org/10.1080/07391102.2023.2188419
- Shengchao Zhou, Kun Wang, Ziwei Hu, Tao Chen, Yao Dong, Rongmei Gao, Mengyuan Wu, Yuhuan Li, Xingyue Ji. Design, synthesis, and structure-activity relationships of a novel class of quinazoline derivatives as coronavirus inhibitors. European Journal of Medicinal Chemistry 2023, 261 , 115831. https://doi.org/10.1016/j.ejmech.2023.115831
- Shailima Rampogu, Tae Sung Jung, Min Woo Ha, Keun Woo Lee. Repurposing and computational design of PARP inhibitors as SARS-CoV-2 inhibitors. Scientific Reports 2023, 13
(1)
https://doi.org/10.1038/s41598-023-36342-7
- Sunyu Song, Yeseul Kim, Kiwoong Kwak, Hyeonmin Lee, Hyunjae Park, Young Bong Kim, Hee-Jung Lee, Lin-Woo Kang. The N-terminal peptide of the main protease of SARS-CoV-2, targeting dimer interface, inhibits its proteolytic activity. BMB Reports 2023, 56
(11)
, 606-611. https://doi.org/10.5483/BMBRep.2023-0153
- Jie Liu, Liang Xu, Wenjing Guo, Zoe Li, Md Kamrul Hasan Khan, Weigong Ge, Tucker A Patterson, Huixiao Hong. Developing a SARS-CoV-2 main protease binding prediction random forest model for drug repurposing for COVID-19 treatment. Experimental Biology and Medicine 2023, 82 https://doi.org/10.1177/15353702231209413
- Kun Zhou, Daquan Chen. Conventional Understanding of SARS‐CoV‐2 M
pro
and Common Strategies for Developing Its Inhibitors. ChemBioChem 2023, 24
(22)
https://doi.org/10.1002/cbic.202300301
- Navid Kaboudi, Nadine Krüger, Maryam Hamzeh‐Mivehroud. Development of novel ligands against SARS‐CoV‐2 M
pro
enzyme: an
in silico
and
in vitro
Study. Molecular Informatics 2023, 42
(11)
https://doi.org/10.1002/minf.202300120
- Wandong Liu, Juan Wang, Suyun Wang, Kairui Yue, Yu Hu, Xiaochun Liu, Lihao Wang, Shengbiao Wan, Ximing Xu. Discovery of new non-covalent and covalent inhibitors targeting SARS-CoV-2 papain-like protease and main protease. Bioorganic Chemistry 2023, 140 , 106830. https://doi.org/10.1016/j.bioorg.2023.106830
- Xin Li, Yongcheng Song. Structure and function of SARS-CoV and SARS-CoV-2 main proteases and their inhibition: A comprehensive review. European Journal of Medicinal Chemistry 2023, 260 , 115772. https://doi.org/10.1016/j.ejmech.2023.115772
- Jiaojiao Li, Lin Zhu, Zheng Qin, Zhengfu Li, Xun Gao, Jing Ji, Jinyang Shen. Discovery of Potential Compounds Against SARS-CoV-2 Based on
3CLpro/RdRp Dual-target: An In silico Approach. Letters in Drug Design & Discovery 2023, 20
(11)
, 1846-1860. https://doi.org/10.2174/1570180819666220818145647
- Yinkai Duan, Haofeng Wang, Zhenghong Yuan, Haitao Yang. Structural biology of SARS-CoV-2 Mpro and drug discovery. Current Opinion in Structural Biology 2023, 82 , 102667. https://doi.org/10.1016/j.sbi.2023.102667
- Xiaojing Pang, Wei Xu, Yang Liu, Hua Li, Lixia Chen. The research progress of SARS-CoV-2 main protease inhibitors from 2020 to 2022. European Journal of Medicinal Chemistry 2023, 257 , 115491. https://doi.org/10.1016/j.ejmech.2023.115491
- Sabikeh Azimi, Muna S. Merza, Fatemeh Ghasemi, Hasan Ali Dhahi, Farid Baradarbarjastehbaf, Mehdi Moosavi, Pouya Ghamari Kargar, Christophe Len. Green and rapid and instrumental one-pot method for the synthesis of imidazolines having potential anti-SARS-CoV-2 main protease activity. Sustainable Chemistry and Pharmacy 2023, 34 , 101136. https://doi.org/10.1016/j.scp.2023.101136
- Andrea Citarella, Alessandro Dimasi, Davide Moi, Daniele Passarella, Angela Scala, Anna Piperno, Nicola Micale. Recent Advances in SARS-CoV-2 Main Protease Inhibitors: From Nirmatrelvir to Future Perspectives. Biomolecules 2023, 13
(9)
, 1339. https://doi.org/10.3390/biom13091339
- Amanda Resende Tucci, Raquel Mello da Rosa, Alice Santos Rosa, Otávio Augusto Chaves, Vivian Neuza Santos Ferreira, Thamara Kelcya Fonseca Oliveira, Daniel Dias Coutinho Souza, Nathalia Roberto Resende Borba, Luciano Dornelles, Nayra Salazar Rocha, João Candido Pilar Mayer, João B. Teixeira da Rocha, Oscar Endrigo D. Rodrigues, Milene Dias Miranda. Antiviral Effect of 5′-Arylchalcogeno-3-aminothymidine Derivatives in SARS-CoV-2 Infection. Molecules 2023, 28
(18)
, 6696. https://doi.org/10.3390/molecules28186696
- Tushar Saha, Shubhra Sinha, Rhodri Harfoot, Miguel E. Quiñones-Mateu, Shyamal C. Das. Spray-Dried Inhalable Microparticles Combining Remdesivir and Ebselen against SARS-CoV-2 Infection. Pharmaceutics 2023, 15
(9)
, 2229. https://doi.org/10.3390/pharmaceutics15092229
- Swaraj Mohanty, Soumya Lipsa Rath, Poornima Sharma, Yasmin Ahmad. An Evidence of Drug Repurposing for COVID-19 Pandemic Based on In silico Investigation from Phenolic Derivatives of Silybum Marianum Against SARS-Cov-2 Proteins. OBM Genetics 2023, 07
(03)
, 1-17. https://doi.org/10.21926/obm.genet.2303186
- Guilherme Wildner, Amanda Resende Tucci, Alessandro de Souza Prestes, Talise Muller, Alice dos Santos Rosa, Nathalia Roberto R. Borba, Vivian Neuza Ferreira, João Batista Teixeira Rocha, Milene Dias Miranda, Nilda Vargas Barbosa. Ebselen and Diphenyl Diselenide Inhibit SARS-CoV-2 Replication at Non-Toxic Concentrations to Human Cell Lines. Vaccines 2023, 11
(7)
, 1222. https://doi.org/10.3390/vaccines11071222
- Hossein Mousavi, Behzad Zeynizadeh, Mehdi Rimaz. Green and efficient one-pot three-component synthesis of novel drug-like furo[2,3-d]pyrimidines as potential active site inhibitors and putative allosteric hotspots modulators of both SARS-CoV-2 MPro and PLPro. Bioorganic Chemistry 2023, 135 , 106390. https://doi.org/10.1016/j.bioorg.2023.106390
- Chamandi S. Dampalla, Matthew J. Miller, Yunjeong Kim, Alexandria Zabiegala, Harry Nhat Nguyen, Trent K. Madden, Hayden A. Thurman, Alexandra J. Machen, Anne Cooper, Lijun Liu, Kevin P. Battaile, Scott Lovell, Kyeong-Ok Chang, William C. Groutas. Structure-guided design of direct-acting antivirals that exploit the gem-dimethyl effect and potently inhibit 3CL proteases of severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) and middle east respiratory syndrome coronavirus (MERS-CoV). European Journal of Medicinal Chemistry 2023, 254 , 115376. https://doi.org/10.1016/j.ejmech.2023.115376
- Samson Afewerki, Thiago Domingues Stocco, André Diniz Rosa da Silva, André Sales Aguiar Furtado, Gustavo Fernandes de Sousa, Guillermo U. Ruiz-Esparza, Thomas J. Webster, Fernanda R. Marciano, Maria Strømme, Yu Shrike Zhang, Anderson Oliveira Lobo. In vitro high-content tissue models to address precision medicine challenges. Molecular Aspects of Medicine 2023, 91 , 101108. https://doi.org/10.1016/j.mam.2022.101108
- J. Jeya Vandana, Cassandra Manrique, Lauretta A. Lacko, Shuibing Chen. Human pluripotent-stem-cell-derived organoids for drug discovery and evaluation. Cell Stem Cell 2023, 30
(5)
, 571-591. https://doi.org/10.1016/j.stem.2023.04.011
- Xuming Tang, Dongxiang Xue, Tuo Zhang, Benjamin E. Nilsson-Payant, Lucia Carrau, Xiaohua Duan, Miriam Gordillo, Adrian Y. Tan, Yunping Qiu, Jenny Xiang, Robert E. Schwartz, Benjamin R. tenOever, Todd Evans, Shuibing Chen. A multi-organoid platform identifies CIART as a key factor for SARS-CoV-2 infection. Nature Cell Biology 2023, 25
(3)
, 381-389. https://doi.org/10.1038/s41556-023-01095-y
- Matteo Pavan, Stefano Moro. Lessons Learnt from COVID-19: Computational Strategies for Facing Present and Future Pandemics. International Journal of Molecular Sciences 2023, 24
(5)
, 4401. https://doi.org/10.3390/ijms24054401
- Luca Sancineto, Vanessa Nascimento. Chalcogen-containing Therapeutic Molecules. 2023, 592-602. https://doi.org/10.1039/BK9781839167386-00592
- Hongtao Xu, Wei Hou. Selenium-containing heterocycles. 2023, 915-930. https://doi.org/10.1016/B978-0-443-18611-0.00006-1
- Nasim Bakhtiyari, Sepideh Parvizpour, Yosef Masoudi-Sobhanzadeh, Ali Masoudi-Nejad. Computational-based strategies to deal with liver injuries induced by the repurposed drugs against SARS-CoV-2. Informatics in Medicine Unlocked 2023, 41 , 101336. https://doi.org/10.1016/j.imu.2023.101336
- Yong-Ming Yan, Xin Shen, Yan-Peng Li, Yong-Kai Cao, Jiao-Jiao Zhang, Yan Wang, Yong-Xian Cheng. Discovery of anti-SARS-CoV-2 agents from 38 Chinese patent drugs toward respiratory diseases <i>via</i> docking screening. Medicinal Plant Biology 2023, 2
(1)
, 0-0. https://doi.org/10.48130/MPB-2023-0009
- Jiaojiao Zhang, Xin Shen, Yanpeng Li, Yongming Yan, Yan Wang, Yongxian Cheng. Discovery of anti-SARS-CoV-2 agents from commercially available flavor <i>via</i> docking screening. Medicinal Plant Biology 2023, 2
(1)
, 0-0. https://doi.org/10.48130/MPB-2023-0010
- Kai Ni, Bo Che, Chongxin Yang, Youyuan Qin, Rong Gu, Chunhong Wang, Mingzhi Luo, Linhong Deng. Emerging toolset of three-dimensional pulmonary cell culture models for simulating lung pathophysiology towards mechanistic elucidation and therapeutic treatment of SARS-COV-2 infection. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.1033043
- Divya M. Teli, Bansari Patel, Mahesh T. Chhabria. Fragment-based design of SARS-CoV-2 Mpro inhibitors. Structural Chemistry 2022, 33
(6)
, 2155-2168. https://doi.org/10.1007/s11224-022-02031-w
- Mahima Chauhan, Vijay Kumar Bhardwaj, Asheesh Kumar, Vinod Kumar, Pawan Kumar, M. Ghalib Enayathullah, Jessie Thomas, Joel George, Bokara Kiran Kumar, Rituraj Purohit, Arun Kumar, Sanjay Kumar. Theaflavin 3-gallate inhibits the main protease (Mpro) of SARS-CoV-2 and reduces its count in vitro. Scientific Reports 2022, 12
(1)
https://doi.org/10.1038/s41598-022-17558-5
- Kunal Nepali, Ram Sharma, Sachin Sharma, Amandeep Thakur, Jing-Ping Liou. Beyond the vaccines: a glance at the small molecule and peptide-based anti-COVID19 arsenal. Journal of Biomedical Science 2022, 29
(1)
https://doi.org/10.1186/s12929-022-00847-6
- Vijayan Ramachandran, Yanyun Liu, Qianying He, Andrew Tang, Patrick Ronaldson, Dominik Schenten, Rui Chang. Novel inhibitors against COVID-19 main protease suppressed viral infection. 2022https://doi.org/10.1101/2022.11.05.515305
- Kirsten McAulay, Alan Bilsland, Marta Bon. Reactivity of Covalent Fragments and Their Role in Fragment Based Drug Discovery. Pharmaceuticals 2022, 15
(11)
, 1366. https://doi.org/10.3390/ph15111366
- Suhasini M. Iyengar, Kelly K. Barnsley, Hoang Yen Vu, Ian Jef A. Bongalonta, Alyssa S. Herrod, Jasmine A. Scott, Mary Jo Ondrechen. Identification and characterization of alternative sites and molecular probes for SARS-CoV-2 target proteins. Frontiers in Chemistry 2022, 10 https://doi.org/10.3389/fchem.2022.1017394
- Pushyaraga P. Venugopal, Omkar Singh, Debashree Chakraborty. Understanding the role of water on temperature-dependent structural modifications of SARS CoV-2 main protease binding sites. Journal of Molecular Liquids 2022, 363 , 119867. https://doi.org/10.1016/j.molliq.2022.119867
- Ly Hien Doan, Li-Wei Chu, Zi-Yi Huang, Anh Thuc Nguyen, Chia-Yin Lee, Chien-Ling Huang, Yu-Fen Chang, Wen-Yu Hsieh, Trang Thi Huyen Nguyen, Chao-Hsiung Lin, Chun-Li Su, Tsung-Hsien Chuang, Jin-Mei Lai, Feng-Sheng Wang, Chia-Jui Yang, Hui-Kang Liu, Yueh-Hsin Ping, Chi-Ying F. Huang. Virofree, an Herbal Medicine-Based Formula, Interrupts the Viral Infection of Delta and Omicron Variants of SARS-CoV-2. Frontiers in Pharmacology 2022, 13 https://doi.org/10.3389/fphar.2022.905197
- Nicola Gambacorta, Leonardo Caputo, Laura Quintieri, Linda Monaci, Fulvio Ciriaco, Orazio Nicolotti. Rational Discovery of Antiviral Whey Protein-Derived Small Peptides Targeting the SARS-CoV-2 Main Protease. Biomedicines 2022, 10
(5)
, 1067. https://doi.org/10.3390/biomedicines10051067
- Supaphorn Seetaha, Phatcharin Khamplong, Panatda Wanaragthai, Thitinan Aiebchun, Siriluk Ratanabunyong, Sucheewin Krobthong, Yodying Yingchutrakul, Jatuporn Rattanasrisomporn, Kiattawee Choowongkomon. KERRA, Mixed Medicinal Plant Extracts, Inhibits SARS-CoV-2 Targets Enzymes and Feline Coronavirus. COVID 2022, 2
(5)
, 621-632. https://doi.org/10.3390/covid2050046
- Zhonglei Wang, Liyan Yang. Broad‐spectrum prodrugs with anti‐SARS‐CoV‐2 activities: Strategies, benefits, and challenges. Journal of Medical Virology 2022, 94
(4)
, 1373-1390. https://doi.org/10.1002/jmv.27517
- Yuling Han, Liuliu Yang, Lauretta A. Lacko, Shuibing Chen. Human organoid models to study SARS-CoV-2 infection. Nature Methods 2022, 19
(4)
, 418-428. https://doi.org/10.1038/s41592-022-01453-y
- Guillem Macip, Pol Garcia-Segura, Júlia Mestres-Truyol, Bryan Saldivar-Espinoza, Gerard Pujadas, Santiago Garcia-Vallvé. A Review of the Current Landscape of SARS-CoV-2 Main Protease Inhibitors: Have We Hit the Bullseye Yet?. International Journal of Molecular Sciences 2022, 23
(1)
, 259. https://doi.org/10.3390/ijms23010259
- Prasat Kittakoop, Dhanushka Darshana, Rapeepat Sangsuwan, Chulabhorn Mahidol. Alkaloids and Alkaloid-Like Compounds are Potential Scaffolds of Antiviral Agents against SARS-CoV-2 (COVID-19) Virus. HETEROCYCLES 2022, 105
(1)
, 115. https://doi.org/10.3987/REV-22-SR(R)3
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 50 other publications.
- 1Li, F.; Li, W.; Farzan, M.; Harrison, S. C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309 (5742), 1864– 1868, DOI: 10.1126/science.11164801https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpvFCisLw%253D&md5=0a7ac2fb7a76c97979d859c0489c682fStructure of SARS Coronavirus Spike Receptor-Binding Domain Complexed with ReceptorLi, Fang; Li, Wenhui; Farzan, Michael; Harrison, Stephen C.Science (Washington, DC, United States) (2005), 309 (5742), 1864-1868CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The spike protein (S) of SARS coronavirus (SARS-CoV) attaches the virus to its cellular receptor, angiotensin-converting enzyme 2 (ACE2). A defined receptor-binding domain (RBD) on S mediates this interaction. The crystal structure at 2.9 angstrom resoln. of the RBD bound with the peptidase domain of human ACE2 shows that the RBD presents a gently concave surface, which cradles the N-terminal lobe of the peptidase. The at. details at the interface between the two proteins clarify the importance of residue changes that facilitate efficient cross-species infection and human-to-human transmission. The structure of the RBD suggests ways to make truncated disulfide-stabilized RBD variants for use in the design of coronavirus vaccines.
- 2Phan, T. Novel coronavirus: From discovery to clinical diagnostics. Infect., Genet. Evol. 2020, 79, 104211, DOI: 10.1016/j.meegid.2020.1042112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFOit7g%253D&md5=3cb3ef734c4f4f85479d8b55a2daf94cNovel coronavirus: From discovery to clinical diagnosticsPhan, TungInfection, Genetics and Evolution (2020), 79 (), 104211CODEN: IGENCN; ISSN:1567-1348. (Elsevier B.V.)A novel coronavirus designated as 2019-nCoV first appeared in Wuhan, China in late Dec. 2019. Dozens of people died in China, and thousands of people infected as 2019-nCoV continues to spread around the world. We have described the discovery, emergence, genomic characteristics, and clin. diagnostics of 2019-nCoV.
- 3Pillaiyar, T.; Manickam, M.; Namasivayam, V.; Hayashi, Y.; Jung, S. H. An Overview of Severe Acute Respiratory Syndrome-Coronavirus (SARS-CoV) 3CL Protease Inhibitors: Peptidomimetics and Small Molecule Chemotherapy. J. Med. Chem. 2016, 59 (14), 6595– 628, DOI: 10.1021/acs.jmedchem.5b014613https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XisVyns7c%253D&md5=6ce37076204280f64a7f362633fcee1aAn Overview of Severe Acute Respiratory Syndrome-Coronavirus (SARS-CoV) 3CL Protease Inhibitors: Peptidomimetics and Small Molecule ChemotherapyPillaiyar, Thanigaimalai; Manickam, Manoj; Namasivayam, Vigneshwaran; Hayashi, Yoshio; Jung, Sang-HunJournal of Medicinal Chemistry (2016), 59 (14), 6595-6628CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Severe acute respiratory syndrome (SARS) is caused by a newly emerged coronavirus that infected more than 8000 individuals and resulted in more than 800 (10-15%) fatalities in 2003. The causative agent of SARS has been identified as a novel human coronavirus (SARS-CoV), and its viral protease, SARS-CoV 3CLpro, has been shown to be essential for replication and has hence been recognized as a potent drug target for SARS infection. Currently, there is no effective treatment for this epidemic despite the intensive research that has been undertaken since 2003 (over 3500 publications). This perspective focuses on the status of various efficacious anti-SARS-CoV 3CLpro chemotherapies discovered during the last 12 years (2003-2015) from all sources, including lab. synthetic methods, natural products, and virtual screening. We describe here mainly peptidomimetic and small mol. inhibitors of SARS-CoV 3CLpro. Attempts have been made to provide a complete description of the structural features and binding modes of these inhibitors under many conditions.
- 4Ziebuhr, J.; Snijder, E. J.; Gorbalenya, A. E. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J. Gen. Virol. 2000, 81 (4), 853– 879, DOI: 10.1099/0022-1317-81-4-8534https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXisVektLg%253D&md5=5293084b82548029518d57e401eccc3eVirus-encoded proteinases and proteolytic processing in the nidoviralesZiebuhr, John; Snijder, Eric J.; Gorbalenya, Alexander E.Journal of General Virology (2000), 81 (4), 853-879CODEN: JGVIAY; ISSN:0022-1317. (Society for General Microbiology)A review with 128 refs. The review is organized into five main sections. In the first section nidovirus proteinases are introduced and classified into main and accessory proteinases. In the next two sections a brief overview of two classes of nidovirus proteinases is presented and, in this context, the coronavirus and arterivirus enzymes are compared to each other and to the prototypic proteinases. This is followed by a detailed description of the nidovirus proteinases themselves. The review is concluded by two sections that describe the regulatory role of the proteinases during virus replication and then give an outline of future perspectives concerning nidovirus proteinases.
- 5Báez-Santos, Y. M.; St. John, S. E.; Mesecar, A. D. The SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compounds. Antiviral Res. 2015, 115, 21– 38, DOI: 10.1016/j.antiviral.2014.12.0155https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmtVOnsw%253D%253D&md5=fcdde62e05b499546c73f3014ea521adThe SARS-coronavirus papain-like protease: Structure, function and inhibition by designed antiviral compoundsBaez-Santos, Yahira M.; St. John, Sarah E.; Mesecar, Andrew D.Antiviral Research (2015), 115 (), 21-38CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)A review. Over 10 years have passed since the deadly human coronavirus that causes severe acute respiratory syndrome (SARS-CoV) emerged from the Guangdong Province of China. Despite the fact that the SARS-CoV pandemic infected over 8500 individuals, claimed over 800 lives and cost billions of dollars in economic loss worldwide, there still are no clin. approved antiviral drugs, vaccines or monoclonal antibody therapies to treat SARS-CoV infections. The recent emergence of the deadly human coronavirus that causes Middle East respiratory syndrome (MERS-CoV) is a sobering reminder that new and deadly coronaviruses can emerge at any time with the potential to become pandemics. Therefore, the continued development of therapeutic and prophylactic countermeasures to potentially deadly coronaviruses is warranted. The coronaviral proteases, papain-like protease (PLpro) and 3C-like protease (3CLpro), are attractive antiviral drug targets because they are essential for coronaviral replication. Although the primary function of PLpro and 3CLpro are to process the viral polyprotein in a coordinated manner, PLpro has the addnl. function of stripping ubiquitin and ISG15 from host-cell proteins to aid coronaviruses in their evasion of the host innate immune responses. Therefore, targeting PLpro with antiviral drugs may have an advantage in not only inhibiting viral replication but also inhibiting the dysregulation of signaling cascades in infected cells that may lead to cell death in surrounding, uninfected cells. This review provides an up-to-date discussion on the SARS-CoV papain-like protease including a brief overview of the SARS-CoV genome and replication followed by a more in-depth discussion on the structure and catalytic mechanism of SARS-CoV PLpro, the multiple cellular functions of SARS-CoV PLpro, the inhibition of SARS-CoV PLpro by small mol. inhibitors, and the prospect of inhibiting papain-like protease from other coronaviruses. This paper forms part of a series of invited articles in Antiviral Research on "From SARS to MERS: 10 years of research on highly pathogenic human coronaviruses.".
- 6Lee, T. W.; Cherney, M. M.; Huitema, C.; Liu, J.; James, K. E.; Powers, J. C.; Eltis, L. D.; James, M. N. Crystal structures of the main peptidase from the SARS coronavirus inhibited by a substrate-like aza-peptide epoxide. J. Mol. Biol. 2005, 353 (5), 1137– 1151, DOI: 10.1016/j.jmb.2005.09.0046https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtFKms7nL&md5=ba0e5fbad36bfc3d259c9221d451ef60Crystal Structures of the Main Peptidase from the SARS Coronavirus Inhibited by a Substrate-like Aza-peptide EpoxideLee, Ting-Wai; Cherney, Maia M.; Huitema, Carly; Liu, Jie; James, Karen Ellis; Powers, James C.; Eltis, Lindsay D.; James, Michael N. G.Journal of Molecular Biology (2005), 353 (5), 1137-1151CODEN: JMOBAK; ISSN:0022-2836. (Elsevier B.V.)The main peptidase (Mpro) from the coronavirus (CoV) causing severe acute respiratory syndrome (SARS) is one of the most attractive mol. targets for the development of anti-SARS agents. We report the irreversible inhibition of SARS-CoV Mpro by an aza-peptide epoxide (APE; kinact/Ki=1900(±400) M-1 s-1). The crystal structures of the Mpro:APE complex in the space groups C2 and P212121 revealed the formation of a covalent bond between the catalytic Cys145 Sγ atom of the peptidase and the epoxide C3 atom of the inhibitor, substantiating the mode of action of this class of cysteine-peptidase inhibitors. The aza-peptide component of APE binds in the substrate-binding regions of Mpro in a substrate-like manner, with excellent structural and chem. complementarity. In addn., the crystal structure of unbound Mpro in the space group C2 revealed that the "N-fingers" (N-terminal residues 1 to 7) of both protomers of Mpro are well defined and the substrate-binding regions of both protomers are in the catalytically competent conformation at the crystn. pH of 6.5, contrary to the previously detd. crystal structures of unbound Mpro in the space group P21.
- 7Xu, X.; Liu, Y.; Weiss, S.; Arnold, E.; Sarafianos, S. G.; Ding, J. Molecular model of SARS coronavirus polymerase: implications for biochemical functions and drug design. Nucleic Acids Res. 2003, 31 (24), 7117– 7130, DOI: 10.1093/nar/gkg9167https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXps1Siu7Y%253D&md5=1c63b9eb468d4b067d4efb3136e18e66Molecular model of SARS coronavirus polymerase: implications for biochemical functions and drug designXu, Xiang; Liu, Yunqing; Weiss, Susan; Arnold, Eddy; Sarafianos, Stefan G.; Ding, JianpingNucleic Acids Research (2003), 31 (24), 7117-7130CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The causative agent of severe acute respiratory syndrome (SARS) is a previously unidentified coronavirus, SARS-CoV. The RNA-dependent RNA polymerase (RdRp) of SARS-CoV plays a pivotal role in viral replication and is a potential target for anti-SARS therapy. There is a lack of structural or biochem. data on any coronavirus polymerase. To provide insights into the structure and function of SARS-CoV RdRp, we have located its conserved motifs that are shared by all RdRps, and built a three-dimensional model of the catalytic domain. The structural model permits us to discuss the potential functional roles of the conserved motifs and residues in replication and their potential interactions with inhibitors of related enzymes. We predict the following important structural attributes for potential anti-SARS-CoV RdRp nucleotide analog inhibitors: hydrogen-bonding capability for the 2' and 3' groups of the sugar ring and C3' endo sugar puckering, and the absence of a hydrophobic binding pocket for non-nucleoside analog inhibitors similar to those obsd. in hepatitis C virus RdRp and human immunodeficiency virus type 1 reverse transcriptase. We propose that the clin. obsd. resistance of SARS to ribavirin is probably due to perturbation of the conserved motif A that controls rNTP binding and fidelity of polymn. Our results suggest that designing anti-SARS therapies can benefit from successful experiences in design of other antiviral drugs. This work should also provide guidance for future biochem. expts.
- 8De Clercq, E.; Li, G. Approved Antiviral Drugs over the Past 50 Years. Clin. Microbiol. Rev. 2016, 29 (3), 695– 747, DOI: 10.1128/CMR.00102-158https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2s%252Fos12htA%253D%253D&md5=1d25d961c1c6a72d6bc4634041bbb6feApproved Antiviral Drugs over the Past 50 YearsDe Clercq Erik; Li GuangdiClinical microbiology reviews (2016), 29 (3), 695-747 ISSN:.Since the first antiviral drug, idoxuridine, was approved in 1963, 90 antiviral drugs categorized into 13 functional groups have been formally approved for the treatment of the following 9 human infectious diseases: (i) HIV infections (protease inhibitors, integrase inhibitors, entry inhibitors, nucleoside reverse transcriptase inhibitors, nonnucleoside reverse transcriptase inhibitors, and acyclic nucleoside phosphonate analogues), (ii) hepatitis B virus (HBV) infections (lamivudine, interferons, nucleoside analogues, and acyclic nucleoside phosphonate analogues), (iii) hepatitis C virus (HCV) infections (ribavirin, interferons, NS3/4A protease inhibitors, NS5A inhibitors, and NS5B polymerase inhibitors), (iv) herpesvirus infections (5-substituted 2'-deoxyuridine analogues, entry inhibitors, nucleoside analogues, pyrophosphate analogues, and acyclic guanosine analogues), (v) influenza virus infections (ribavirin, matrix 2 protein inhibitors, RNA polymerase inhibitors, and neuraminidase inhibitors), (vi) human cytomegalovirus infections (acyclic guanosine analogues, acyclic nucleoside phosphonate analogues, pyrophosphate analogues, and oligonucleotides), (vii) varicella-zoster virus infections (acyclic guanosine analogues, nucleoside analogues, 5-substituted 2'-deoxyuridine analogues, and antibodies), (viii) respiratory syncytial virus infections (ribavirin and antibodies), and (ix) external anogenital warts caused by human papillomavirus infections (imiquimod, sinecatechins, and podofilox). Here, we present for the first time a comprehensive overview of antiviral drugs approved over the past 50 years, shedding light on the development of effective antiviral treatments against current and emerging infectious diseases worldwide.
- 9Kanters, S.; Socias, M. E.; Paton, N. I.; Vitoria, M.; Doherty, M.; Ayers, D.; Popoff, E.; Chan, K.; Cooper, D. A.; Wiens, M. O.; Calmy, A.; Ford, N.; Nsanzimana, S.; Mills, E. J. Comparative efficacy and safety of second-line antiretroviral therapy for treatment of HIV/AIDS: A systematic review and network meta-analysis. Lancet HIV 2017, 4 (10), e433– e441, DOI: 10.1016/S2352-3018(17)30109-1There is no corresponding record for this reference.
- 10Deeks, E. D. Darunavir: a review of its use in the management of HIV-1 infection. Drugs 2014, 74 (1), 99– 125, DOI: 10.1007/s40265-013-0159-310https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXptFajuw%253D%253D&md5=b5f8e515fd0c0ce64d97b0c126c18ce4Darunavir: A Review of Its Use in the Management of HIV-1 InfectionDeeks, Emma D.Drugs (2014), 74 (1), 99-125CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. The latest HIV-1 protease inhibitor (PI) darunavir (Prezista®) has a high genetic barrier to resistance development and is active against wild-type HIV and HIV strains no longer susceptible to some older PIs. Ritonavir-boosted darunavir, as a component of antiretroviral therapy (ART), is indicated for the treatment of HIV-1 infection in adult and paediatric patients (aged ≥3 years), with or without treatment experience (details vary depending on region of approval). Several open-label or partially-blinded trials have evaluated the efficacy of ritonavir-boosted darunavir ART regimens for up to 192 wk in these settings. In treatment-naive adults, once-daily boosted darunavir was no less effective in establishing virol. suppression than once- or twice-daily boosted lopinavir, yet was more effective at maintaining suppression long term. Moreover, treatment-experienced adults with no darunavir resistance-assocd. mutations (RAMs) had no less effective viral load suppression with once-daily than with twice-daily boosted darunavir. In treatment-experienced adults, including some with multiple major PI RAMs, twice-daily boosted darunavir was more effective than twice-daily boosted lopinavir or boosted control PIs in reducing viral load, and provided virol. benefit as part of a salvage regimen in those with few remaining treatment options. Boosted darunavir also reduced viral load when administered once-daily in treatment-naive adolescents or twice-daily in treatment-experienced children and adolescents. Boosted darunavir is generally well tolerated, with gastrointestinal disturbances and lipid abnormalities among the most common tolerability issues. It has a lipid profile more favorable than that of boosted lopinavir in terms of total cholesterol and triglyceride changes and, when administered once daily, its lipid effects are generally similar to those of boosted atazanavir. Thus, boosted darunavir is a useful option for the ART regimens of adult and paediatric patients with HIV-1 infection.
- 11la Porte, C. J. Saquinavir, the pioneer antiretroviral protease inhibitor. Expert Opin. Drug Metab. Toxicol. 2009, 5 (10), 1313– 1322, DOI: 10.1517/1742525090327316011https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFCgsrfM&md5=337b9225b09c6cabc0a87f41272b1edeSaquinavir, the pioneer antiretroviral protease inhibitorLa Porte, Charles J. L.Expert Opinion on Drug Metabolism & Toxicology (2009), 5 (10), 1313-1322CODEN: EODMAP; ISSN:1742-5255. (Informa Healthcare)A review. Background: The treatment of HIV infection underwent a major change in 1995 when saquinavir was the first protease inhibitor introduced into the market. This drug made the use of combination therapy in the treatment of HIV possible and increased the success rate of treatment. Objective: This article will review recent literature on saquinavir to define its current role in HIV treatment, among the newer antiretroviral drugs. Methods: Scientific literature and conference presentations were evaluated for relevant information pertaining to saquinavir. Results/conclusions: Although underused, saquinavir has good efficacy and tolerability when compared to other protease inhibitors. The film-coated tablet formulation improved pill burden. Saquinavir still has potential in the treatment of adults, children and pregnant women.
- 12Heo, Y. A.; Deeks, E. D. Sofosbuvir/Velpatasvir/Voxilaprevir: A Review in Chronic Hepatitis C. Drugs 2018, 78 (5), 577– 587, DOI: 10.1007/s40265-018-0895-512https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkslGjt7k%253D&md5=d4557e43a321ca3a9a30d996ac5f77baSofosbuvir/Velpatasvir/Voxilaprevir: A Review in Chronic Hepatitis CHeo, Young-A.; Deeks, Emma D.Drugs (2018), 78 (5), 577-587CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A fixed-dose combination of the hepatitis C virus (HCV) NS5B polymerase inhibitor sofosbuvir, the HCV NS5A inhibitor velpatasvir and the HCV NS3/4A protease inhibitor voxilaprevir (sofosbuvir/velpatasvir/voxilaprevir; Vosevi) is approved in the EU for the treatment of chronic HCV genotype 1, 2, 3, 4, 5 or 6 infection in adults. In the phase III POLARIS trials, in patients who had HCV genotype 1-6 infection with or without compensated cirrhosis, overall rates of sustained virol. response at 12 wk post-treatment (SVR12) with sofosbuvir/velpatasvir/voxilaprevir were high after 8 wk of treatment in direct-acting antiviral (DAA)-naive patients and 12 wk of treatment in DAA-experienced patients. However, 8 wk of sofosbuvir/velpatasvir/voxilaprevir was inferior to 12 wk of sofosbuvir/velpatasvir in cirrhotic or non-cirrhotic DAA-naive patients with HCV genotype 1, 2, 4, 5 or 6 infection and non-cirrhotic DAA-naive patients with HCV genotype 3 infection, mostly due to an insufficient treatment period. Sofosbuvir/velpatasvir/voxilaprevir was generally well tolerated, with most adverse events being of mild or moderate intensity. The most common adverse events included headache, fatigue, nausea and diarrhoea. In conclusion, sofosbuvir/velpatasvir/voxilaprevir is an important and effective option for the treatment of HCV genotype 1-6 infection in adults, esp. those who have previously failed a DAA therapy with or without an HCV NS5A inhibitor.
- 13Sanford, M. Simeprevir: a review of its use in patients with chronic hepatitis C virus infection. Drugs 2015, 75 (2), 183– 196, DOI: 10.1007/s40265-014-0341-213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVCnt7k%253D&md5=20ef9a811518a120a68eb79241f0d735Simeprevir: A Review of Its Use in Patients with Chronic Hepatitis C Virus InfectionSanford, MarkDrugs (2015), 75 (2), 183-196CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. Simeprevir (Olysio; Galexos; Sovriad) is an orally-administered NS3/4A protease inhibitor for use in combined drug regimens against chronic hepatitis C virus (HCV) infection. This article reviews studies relevant to the EU simeprevir label. In proof-of-concept studies, simeprevir had potent antiviral activity against all HCV genotypes, except genotype 3. In trials in patients with chronic HCV genotype 1 infection, week-12 sustained virol. response (SVR12) rates in treatment-naive patients and prior relapsers were significantly higher with simeprevir plus peginterferon-α/ribavirin (PR) [79-89 %] than with placebo plus PR (36-62 %). In prior partial/null responders, the SVR12 rate with simeprevir plus PR (54 %) was noninferior to that with telaprevir plus PR (55 %). Simeprevir plus PR was also efficacious in patients with HCV genotype 1/HIV-1 co-infection. In prior null responders without severe liver fibrosis (cohort 1) and treatment-naive patients with severe liver fibrosis (cohort 2) treated with simeprevir plus sofosbuvir, the SVR12 rate for the two cohorts combined was 92 %. In patients with chronic HCV genotype 4 infection, the SVR12 rates with simeprevir plus PR were 83, 87 and 40 % in treatment-naive patients, prior relapsers and prior null responders, resp. Grade 3-4 adverse event, serious adverse event and treatment withdrawal rates with simeprevir plus PR were similar to those with placebo plus PR. Skin rashes with simeprevir were mostly mild or moderate; serious photosensitivity reactions occur, but are rare. Simeprevir is efficacious and generally well tolerated in patients with chronic HCV genotypes 1 and 4 infection. Studies of simeprevir in interferon-free regimens and in other subpopulations with HCV infections will be of interest.
- 14Chopp, S.; Vanderwall, R.; Hult, A.; Klepser, M. Simeprevir and sofosbuvir for treatment of hepatitis C infection. Am. J. Health-Syst. Pharm. 2015, 72 (17), 1445– 1455, DOI: 10.2146/ajhp14029014https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmtFGgt7k%253D&md5=b8e2a8a629e121eeba1b7bb37a5fa5e6Simeprevir and sofosbuvir for treatment of hepatitis C infectionChopp, Shelby; Vanderwall, Rebecca; Hult, Amanda; Klepser, MichaelAmerican Journal of Health-System Pharmacy (2015), 72 (17), 1445-1455CODEN: AHSPEK; ISSN:1079-2082. (American Society of Health-System Pharmacists)Purpose. The pharmacol., pharmacokinetics, efficacy, safety, costs, and place in therapy of simeprevir and sofosbuvir in the management of hepatitis C virus (HCV) infection are reviewed. Summary. Sofosbuvir and simeprevir are classified as direct-acting agents because they target specific proteins essential to the replication of HCV. Phase III trials demonstrated that simeprevir in combination with peginterferon alfa and ribavirin was superior to placebo combined with peginterferon alfa and ribavirin in achieving a sustained virol. response in both treatment-naive patients and patients who relapsed after treatment with peginterferon alfa-2a or alfa-2b and ribavirin. Q80K polymorphism substantially decreases the efficacy of simeprevir. Clin. trials revealed that sofosbuvir in combination with ribavirin was superior to peginterferon plus ribavirin against HCV genotype 2 infection and as effective as peginterferon plus ribavirin against HCV genotype 3 infection. These findings were significant because they demonstrated the effectiveness of an anti-HCV regimen that did not include peginterferon alfa. Sofosbuvir has much better adverse-effect and drug interaction profiles than previous hepatitis C antiviral agents. Both simeprevir and sofosbuvir are approved for the treatment of chronic hepatitis C in combination with other antiviral medications. Simeprevir has been approved specifically for patients infected with HCV genotype 1 with compensated liver disease (including cirrhosis) in combination with peginterferon alfa-2a or alfa-2b and ribavirin. Sofosbuvir has shown efficacy in HCV genotypes 1-4. Conclusion. Simeprevir and sofasbuvir have advantages in response rates and convenient dosage forms and frequency compared with other HCV treatments; however, they are more expensive than previous HCV therapies.
- 15Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; Duan, Y.; Yu, J.; Wang, L.; Yang, K.; Liu, F.; Jiang, R.; Yang, X.; You, T.; Liu, X.; Yang, X.; Bai, F.; Liu, H.; Liu, X.; Guddat, L. W.; Xu, W.; Xiao, G.; Qin, C.; Shi, Z.; Jiang, H.; Rao, Z.; Yang, H. Structure of M(pro) from COVID-19 virus and discovery of its inhibitors. Nature 2020, 582, 289– 293, DOI: 10.1038/s41586-020-2223-y15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVyhsrrO&md5=b84f350fe9ce1109485df6caf814ba82Structure of Mpro from SARS-CoV-2 and discovery of its inhibitorsJin, Zhenming; Du, Xiaoyu; Xu, Yechun; Deng, Yongqiang; Liu, Meiqin; Zhao, Yao; Zhang, Bing; Li, Xiaofeng; Zhang, Leike; Peng, Chao; Duan, Yinkai; Yu, Jing; Wang, Lin; Yang, Kailin; Liu, Fengjiang; Jiang, Rendi; Yang, Xinglou; You, Tian; Liu, Xiaoce; Yang, Xiuna; Bai, Fang; Liu, Hong; Liu, Xiang; Guddat, Luke W.; Xu, Wenqing; Xiao, Gengfu; Qin, Chengfeng; Shi, Zhengli; Jiang, Hualiang; Rao, Zihe; Yang, HaitaoNature (London, United Kingdom) (2020), 582 (7811), 289-293CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new coronavirus, known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the etiol. agent responsible for the 2019-2020 viral pneumonia outbreak of coronavirus disease 2019 (COVID-19). Currently, there are no targeted therapeutic agents for the treatment of this disease, and effective treatment options remain very limited. Here, we describe the results of a program that aimed to rapidly discover lead compds. for clin. use, by combining structure-assisted drug design, virtual drug screening and high-throughput screening. This program focused on identifying drug leads that target main protease (Mpro) of SARS-CoV-2: Mpro is a key enzyme of coronaviruses and has a pivotal role in mediating viral replication and transcription, making it an attractive drug target for SARS-CoV-2. We identified a mechanism-based inhibitor (N3) by computer-aided drug design, and then detd. the crystal structure of Mpro of SARS-CoV-2 in complex with this compd. Through a combination of structure-based virtual and high-throughput screening, we assayed more than 10,000 compds.-including approved drugs, drug candidates in clin. trials and other pharmacol. active compds.-as inhibitors of Mpro. Six of these compds. inhibited Mpro, showing half-maximal inhibitory concn. values that ranged from 0.67 to 21.4μM. One of these compds. (ebselen) also exhibited promising antiviral activity in cell-based assays. Our results demonstrate the efficacy of our screening strategy, which can lead to the rapid discovery of drug leads with clin. potential in response to new infectious diseases for which no specific drugs or vaccines are available.
- 16Dai, W.; Zhang, B.; Jiang, X. M.; Su, H.; Li, J.; Zhao, Y.; Xie, X.; Jin, Z.; Peng, J.; Liu, F.; Li, C.; Li, Y.; Bai, F.; Wang, H.; Cheng, X.; Cen, X.; Hu, S.; Yang, X.; Wang, J.; Liu, X.; Xiao, G.; Jiang, H.; Rao, Z.; Zhang, L. K.; Xu, Y.; Yang, H.; Liu, H. Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease. Science 2020, 368 (6497), 1331– 1335, DOI: 10.1126/science.abb448916https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXht1alsrzL&md5=ec87c340410edb919d2584a2b4d33e1aStructure-based design of antiviral drug candidates targeting the SARS-CoV-2 main proteaseDai, Wenhao; Zhang, Bing; Jiang, Xia-Ming; Su, Haixia; Li, Jian; Zhao, Yao; Xie, Xiong; Jin, Zhenming; Peng, Jingjing; Liu, Fengjiang; Li, Chunpu; Li, You; Bai, Fang; Wang, Haofeng; Cheng, Xi; Cen, Xiaobo; Hu, Shulei; Yang, Xiuna; Wang, Jiang; Liu, Xiang; Xiao, Gengfu; Jiang, Hualiang; Rao, Zihe; Zhang, Lei-Ke; Xu, Yechun; Yang, Haitao; Liu, HongScience (Washington, DC, United States) (2020), 368 (6497), 1331-1335CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is the etiol. agent responsible for the global COVID-19 (coronavirus disease 2019) outbreak. The main protease of SARS-CoV-2, Mpro, is a key enzyme that plays a pivotal role in mediating viral replication and transcription. We designed and synthesized two lead compds. (11a and 11b) targeting Mpro. Both exhibited excellent inhibitory activity and potent anti-SARS-CoV-2 infection activity. The x-ray crystal structures of SARS-CoV-2 Mpro in complex with 11a or 11b, both detd. at a resoln. of 1.5 angstroms, showed that the aldehyde groups of 11a and 11b are covalently bound to cysteine 145 of Mpro. Both compds. showed good pharmacokinetic properties in vivo, and 11a also exhibited low toxicity, which suggests that these compds. are promising drug candidates.
- 17Vuong, W.; Khan, M. B.; Fischer, C.; Arutyunova, E.; Lamer, T.; Shields, J.; Saffran, H. A.; McKay, R. T.; van Belkum, M. J.; Joyce, M. A.; Young, H. S.; Tyrrell, D. L.; Vederas, J. C.; Lemieux, M. J. Feline coronavirus drug inhibits the main protease of SARS-CoV-2 and blocks virus replication. Nat. Commun. 2020, 11 (1), 4282, DOI: 10.1038/s41467-020-18096-217https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslWqtbzN&md5=c06d2dc9a65ae289505fe120d41d8885Feline coronavirus drug inhibits the main protease of SARS-CoV-2 and blocks virus replicationVuong, Wayne; Khan, Muhammad Bashir; Fischer, Conrad; Arutyunova, Elena; Lamer, Tess; Shields, Justin; Saffran, Holly A.; McKay, Ryan T.; van Belkum, Marco J.; Joyce, Michael A.; Young, Howard S.; Tyrrell, D. Lorne; Vederas, John C.; Lemieux, M. JoanneNature Communications (2020), 11 (1), 4282CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The main protease, Mpro (or 3CLpro) in SARS-CoV-2 is a viable drug target because of its essential role in the cleavage of the virus polypeptide. Feline infectious peritonitis, a fatal coronavirus infection in cats, was successfully treated previously with a prodrug GC376, a dipeptide-based protease inhibitor. We show the prodrug and its parent GC373, are effective inhibitors of the Mpro from both SARS-CoV and SARS-CoV-2 with IC50 values in the nanomolar range. Crystal structures of SARS-CoV-2 Mpro with these inhibitors have a covalent modification of the nucleophilic Cys145. NMR anal. reveals that inhibition proceeds via reversible formation of a hemithioacetal. GC373 and GC376 are potent inhibitors of SARS-CoV-2 replication in cell culture. They are strong drug candidates for the treatment of human coronavirus infections because they have already been successful in animals. The work here lays the framework for their use in human trials for the treatment of COVID-19.
- 18Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved alpha-ketoamide inhibitors. Science 2020, 368 (6489), 409– 412, DOI: 10.1126/science.abb340518https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnslKrtL8%253D&md5=9ac417c20f54c3327f9de9088b512d52Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitorsZhang, Linlin; Lin, Daizong; Sun, Xinyuanyuan; Curth, Ute; Drosten, Christian; Sauerhering, Lucie; Becker, Stephan; Rox, Katharina; Hilgenfeld, RolfScience (Washington, DC, United States) (2020), 368 (6489), 409-412CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is a global health emergency. An attractive drug target among coronaviruses is the main protease (Mpro, also called 3CLpro) because of its essential role in processing the polyproteins that are translated from the viral RNA. We report the x-ray structures of the unliganded SARS-CoV-2 Mpro and its complex with an α-ketoamide inhibitor. This was derived from a previously designed inhibitor but with the P3-P2 amide bond incorporated into a pyridone ring to enhance the half-life of the compd. in plasma. On the basis of the unliganded structure, we developed the lead compd. into a potent inhibitor of the SARS-CoV-2 Mpro. The pharmacokinetic characterization of the optimized inhibitor reveals a pronounced lung tropism and suitability for administration by the inhalative route.
- 19Jin, Z.; Zhao, Y.; Sun, Y.; Zhang, B.; Wang, H.; Wu, Y.; Zhu, Y.; Zhu, C.; Hu, T.; Du, X.; Duan, Y.; Yu, J.; Yang, X.; Yang, X.; Yang, K.; Liu, X.; Guddat, L. W.; Xiao, G.; Zhang, L.; Yang, H.; Rao, Z. Structural basis for the inhibition of SARS-CoV-2 main protease by antineoplastic drug carmofur. Nat. Struct. Mol. Biol. 2020, 27 (6), 529– 532, DOI: 10.1038/s41594-020-0440-619https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXovVOrtLY%253D&md5=28c7bed4e4154935e6909a53015bf64cStructural basis for the inhibition of SARS-CoV-2 main protease by antineoplastic drug carmofurJin, Zhenming; Zhao, Yao; Sun, Yuan; Zhang, Bing; Wang, Haofeng; Wu, Yan; Zhu, Yan; Zhu, Chen; Hu, Tianyu; Du, Xiaoyu; Duan, Yinkai; Yu, Jing; Yang, Xiaobao; Yang, Xiuna; Yang, Kailin; Liu, Xiang; Guddat, Luke W.; Xiao, Gengfu; Zhang, Leike; Yang, Haitao; Rao, ZiheNature Structural & Molecular Biology (2020), 27 (6), 529-532CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)Abstr.: The antineoplastic drug carmofur is shown to inhibit the SARS-CoV-2 main protease (Mpro). Here, the X-ray crystal structure of Mpro in complex with carmofur reveals that the carbonyl reactive group of carmofur is covalently bound to catalytic Cys145, whereas its fatty acid tail occupies the hydrophobic S2 subsite. Carmofur inhibits viral replication in cells (EC50 = 24.30μM) and is a promising lead compd. to develop new antiviral treatment for COVID-19.
- 20Fu, L.; Ye, F.; Feng, Y.; Yu, F.; Wang, Q.; Wu, Y.; Zhao, C.; Sun, H.; Huang, B.; Niu, P.; Song, H.; Shi, Y.; Li, X.; Tan, W.; Qi, J.; Gao, G. F. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat. Commun. 2020, 11 (1), 4417, DOI: 10.1038/s41467-020-18233-x20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVSmsr3F&md5=273b11d16241499f15103885be398b23Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main proteaseFu, Lifeng; Ye, Fei; Feng, Yong; Yu, Feng; Wang, Qisheng; Wu, Yan; Zhao, Cheng; Sun, Huan; Huang, Baoying; Niu, Peihua; Song, Hao; Shi, Yi; Li, Xuebing; Tan, Wenjie; Qi, Jianxun; Gao, George FuNature Communications (2020), 11 (1), 4417CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: COVID-19 was declared a pandemic on March 11 by WHO, due to its great threat to global public health. The coronavirus main protease (Mpro, also called 3CLpro) is essential for processing and maturation of the viral polyprotein, therefore recognized as an attractive drug target. Here we show that a clin. approved anti-HCV drug, Boceprevir, and a pre-clin. inhibitor against feline infectious peritonitis (corona) virus (FIPV), GC376, both efficaciously inhibit SARS-CoV-2 in Vero cells by targeting Mpro. Moreover, combined application of GC376 with Remdesivir, a nucleotide analog that inhibits viral RNA dependent RNA polymerase (RdRp), results in sterilizing additive effect. Further structural anal. reveals binding of both inhibitors to the catalytically active side of SARS-CoV-2 protease Mpro as main mechanism of inhibition. Our findings may provide crit. information for the optimization and design of more potent inhibitors against the emerging SARS-CoV-2 virus.
- 21Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J. Med. Chem. 2004, 47 (7), 1739– 1749, DOI: 10.1021/jm030643021https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit74%253D&md5=8cc2f0022318b12dd972e9c493375bf9Glide: A new approach for rapid, accurate docking and scoring. 1. method and assessment of docking accuracyFriesner, Richard A.; Banks, Jay L.; Murphy, Robert B.; Halgren, Thomas A.; Klicic, Jasna J.; Mainz, Daniel T.; Repasky, Matthew P.; Knoll, Eric H.; Shelley, Mee; Perry, Jason K.; Shaw, David E.; Francis, Perry; Shenkin, Peter S.Journal of Medicinal Chemistry (2004), 47 (7), 1739-1749CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Unlike other methods for docking ligands to the rigid 3D structure of a known protein receptor, Glide approximates a complete systematic search of the conformational, orientational, and positional space of the docked ligand. In this search, an initial rough positioning and scoring phase that dramatically narrows the search space is followed by torsionally flexible energy optimization on an OPLS-AA nonbonded potential grid for a few hundred surviving candidate poses. The very best candidates are further refined via a Monte Carlo sampling of pose conformation; in some cases, this is crucial to obtaining an accurate docked pose. Selection of the best docked pose uses a model energy function that combines empirical and force-field-based terms. Docking accuracy is assessed by redocking ligands from 282 cocrystd. PDB complexes starting from conformationally optimized ligand geometries that bear no memory of the correctly docked pose. Errors in geometry for the top-ranked pose are less than 1 Å in nearly half of the cases and are greater than 2 Å in only about one-third of them. Comparisons to published data on rms deviations show that Glide is nearly twice as accurate as GOLD and more than twice as accurate as FlexX for ligands having up to 20 rotatable bonds. Glide is also found to be more accurate than the recently described Surflex method.
- 22Halgren, T. A.; Murphy, R. B.; Friesner, R. A.; Beard, H. S.; Frye, L. L.; Pollard, W. T.; Banks, J. L. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. J. Med. Chem. 2004, 47 (7), 1750– 1759, DOI: 10.1021/jm030644s22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsFyit78%253D&md5=33d68dd968e65626b449df61e44e37beGlide: A new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screeningHalgren, Thomas A.; Murphy, Robert B.; Friesner, Richard A.; Beard, Hege S.; Frye, Leah L.; Pollard, W. Thomas; Banks, Jay L.Journal of Medicinal Chemistry (2004), 47 (7), 1750-1759CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Glide's ability to identify active compds. in a database screen is characterized by applying Glide to a diverse set of nine protein receptors. In many cases, two, or even three, protein sites are employed to probe the sensitivity of the results to the site geometry. To make the database screens as realistic as possible, the screens use sets of "druglike" decoy ligands that have been selected to be representative of what we believe is likely to be found in the compd. collection of a pharmaceutical or biotechnol. company. Results are presented for releases 1.8, 2.0, and 2.5 of Glide. The comparisons show that av. measures for both "early" and "global" enrichment for Glide 2.5 are 3 times higher than for Glide 1.8 and more than 2 times higher than for Glide 2.0 because of better results for the least well-handled screens. This improvement in enrichment stems largely from the better balance of the more widely parametrized GlideScore 2.5 function and the inclusion of terms that penalize ligand-protein interactions that violate established principles of phys. chem., particularly as it concerns the exposure to solvent of charged protein and ligand groups. Comparisons to results for the thymidine kinase and estrogen receptors published by Rognan and co-workers (J. Med. Chem. 2000, 43, 4759-4767) show that Glide 2.5 performs better than GOLD 1.1, FlexX 1.8, or DOCK 4.01.
- 23Friesner, R. A.; Murphy, R. B.; Repasky, M. P.; Frye, L. L.; Greenwood, J. R.; Halgren, T. A.; Sanschagrin, P. C.; Mainz, D. T. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J. Med. Chem. 2006, 49 (21), 6177– 6196, DOI: 10.1021/jm051256o23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpvVGmurg%253D&md5=ea428c82ead0d8c27f8c1a7b694a1edfExtra Precision Glide: Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein-Ligand ComplexesFriesner, Richard A.; Murphy, Robert B.; Repasky, Matthew P.; Frye, Leah L.; Greenwood, Jeremy R.; Halgren, Thomas A.; Sanschagrin, Paul C.; Mainz, Daniel T.Journal of Medicinal Chemistry (2006), 49 (21), 6177-6196CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A novel scoring function to est. protein-ligand binding affinities has been developed and implemented as the Glide 4.0 XP scoring function and docking protocol. In addn. to unique water desolvation energy terms, protein-ligand structural motifs leading to enhanced binding affinity are included:(1) hydrophobic enclosure where groups of lipophilic ligand atoms are enclosed on opposite faces by lipophilic protein atoms, (2) neutral-neutral single or correlated hydrogen bonds in a hydrophobically enclosed environment, and (3) five categories of charged-charged hydrogen bonds. The XP scoring function and docking protocol have been developed to reproduce exptl. binding affinities for a set of 198 complexes (RMSDs of 2.26 and 1.73 kcal/mol over all and well-docked ligands, resp.) and to yield quality enrichments for a set of fifteen screens of pharmaceutical importance. Enrichment results demonstrate the importance of the novel XP mol. recognition and water scoring in sepg. active and inactive ligands and avoiding false positives.
- 24Amporndanai, K.; Meng, X.; Shang, W.; Jin, Z.; Rogers, M.; Zhao, Y.; Rao, Z.; Liu, Z. J.; Yang, H.; Zhang, L.; O’Neill, P. M.; Samar Hasnain, S. Inhibition mechanism of SARS-CoV-2 main protease by ebselen and its derivatives. Nat. Commun. 2021, 12 (1), 3061, DOI: 10.1038/s41467-021-23313-724https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtFyntbvK&md5=08fe8105502a0bd2aec4879d78280716Inhibition mechanism of SARS-CoV-2 main protease by ebselen and its derivativesAmporndanai, Kangsa; Meng, Xiaoli; Shang, Weijuan; Jin, Zhenmig; Rogers, Michael; Zhao, Yao; Rao, Zihe; Liu, Zhi-Jie; Yang, Haitao; Zhang, Leike; O'Neill, Paul M.; Samar Hasnain, S.Nature Communications (2021), 12 (1), 3061CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: The SARS-CoV-2 pandemic has triggered global efforts to develop therapeutics. The main protease of SARS-CoV-2 (Mpro), crit. for viral replication, is a key target for therapeutic development. An organoselenium drug called ebselen has been demonstrated to have potent Mpro inhibition and antiviral activity. We have examd. the binding modes of ebselen and its deriv. in Mpro via high resoln. co-crystallog. and investigated their chem. reactivity via mass spectrometry. Stronger Mpro inhibition than ebselen and potent ability to rescue infected cells were obsd. for a no. of derivs. A free selenium atom bound with cysteine of catalytic dyad has been revealed in crystallog. structures of Mpro with ebselen and MR6-31-2 suggesting hydrolysis of the enzyme bound organoselenium covalent adduct and formation of a phenolic byproduct, confirmed by mass spectrometry. The target engagement with selenation mechanism of inhibition suggests wider therapeutic applications of these compds. against SARS-CoV-2 and other zoonotic beta-corona viruses.
- 25Muramatsu, T.; Takemoto, C.; Kim, Y. T.; Wang, H.; Nishii, W.; Terada, T.; Shirouzu, M.; Yokoyama, S. SARS-CoV 3CL protease cleaves its C-terminal autoprocessing site by novel subsite cooperativity. Proc. Natl. Acad. Sci. U. S. A. 2016, 113 (46), 12997– 13002, DOI: 10.1073/pnas.160132711325https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslKnu7%252FP&md5=f82749f292da1066e3eb4717dcbc66f2SARS-CoV 3CL protease cleaves its C-terminal autoprocessing site by novel subsite cooperativityMuramatsu, Tomonari; Takemoto, Chie; Kim, Yong-Tae; Wang, Hongfei; Nishii, Wataru; Terada, Takaho; Shirouzu, Mikako; Yokoyama, ShigeyukiProceedings of the National Academy of Sciences of the United States of America (2016), 113 (46), 12997-13002CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The 3C-like protease (3CLpro) of severe acute respiratory syndrome coronavirus (SARS-CoV) cleaves 11 sites in the polyproteins, including its own N- and C-terminal autoprocessing sites, by recognizing P4-P1 and P1'. In this study, we detd. the crystal structure of 3CLpro with the C-terminal prosequence and the catalytic-site C145A mutation, in which the enzyme binds the C-terminal prosequence of another mol. Surprisingly, Phe at the P3' position [Phe(P3')] is snugly accommodated in the S3' pocket. Mutations of Phe(P3') impaired the C-terminal autoprocessing, but did not affect N-terminal autoprocessing. This difference was ascribed to the P2 residue, Phe(P2) and Leu(P2), in the C- and N-terminal sites, as follows. The S3' subsite is formed by Phe(P2)-induced conformational changes of 3CLpro and the direct involvement of Phe(P2) itself. In contrast, the N-terminal prosequence with Leu(P2) does not cause such conformational changes for the S3' subsite formation. In fact, the mutation of Phe(P2) to Leu in the C-terminal autoprocessing site abolishes the dependence on Phe(P3'). These mechanisms explain why Phe is required at the P3' position when the P2 position is occupied by Phe rather than Leu, which reveals a type of subsite cooperativity. Moreover, the peptide consisting of P4-P1 with Leu(P2) inhibits protease activity, whereas that with Phe(P2) exhibits a much smaller inhibitory effect, because Phe(P3') is missing. Thus, this subsite cooperativity likely exists to avoid the autoinhibition of the enzyme by its mature C-terminal sequence, and to retain the efficient C-terminal autoprocessing by the use of Phe(P2).
- 26Sarma, B. K.; Mugesh, G. Antioxidant activity of the anti-inflammatory compound ebselen: a reversible cyclization pathway via selenenic and seleninic acid intermediates. Chem. - Eur. J. 2008, 14 (34), 10603– 10614, DOI: 10.1002/chem.20080125826https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsV2rsLfI&md5=6a82b84de2f8fddc7f2ac90cd62e0b15Antioxidant activity of the anti-inflammatory compound ebselen: a reversible cyclization pathway via selenenic and seleninic acid intermediatesSarma, Bani Kanta; Mugesh, GovindasamyChemistry - A European Journal (2008), 14 (34), 10603-10614CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)A revised mechanism that accounts for the glutathione peroxidase (GPx)-like catalytic activity of the organoselenium compd. ebselen (2-phenyl-1,2-benzisoselenazol-3(2H)-one) is described. The reaction of ebselen with H2O2 yields seleninic acid 2-PhNHC(O)C6H4SeO2H as the only oxidized product. The X- ray crystal structure of the seleninic acid shows that the Se atom is involved in a noncovalent interaction with the carbonyl O atom. In the presence of excess PhSH, the Se-N bond in ebselen is readily cleaved by the thiol to produce the corresponding selenenyl sulfide 2-PhNHC(O)C6H4SeSPh. The selenenyl sulfide thus produced undergoes a disproportionation in the presence of H2O2 to produce the diselenide, which upon reaction with H2O2, produces a mixt. of selenenic and seleninic acids. The addn. of thiol to the mixt. contg. selenenic and seleninic acids gives the selenenyl sulfide. When the concn. of the thiol is relatively low in the reaction mixt., the selenenic acid undergoes a rapid cyclization to produce ebselen. The seleninic acid, however, reacts with the diselenide to produce ebselen as the final product. DFT calcns. show that the cyclization of selenenic acids to the corresponding selenenyl amides is more favored than that of sulfenic acids to the corresponding sulfenyl amides. The regeneration of ebselen under a variety of conditions protects the Se moiety from irreversible inactivation, which may be responsible for the biol. activities of ebselen.
- 27Haenen, G. R.; De Rooij, B. M.; Vermeulen, N. P.; Bast, A. Mechanism of the reaction of ebselen with endogenous thiols: Dihydrolipoate is a better cofactor than glutathione in the peroxidase activity of ebselen. Mol. Pharmacol. 1990, 37 (3), 412– 42227https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXhvFWlsrg%253D&md5=ce9215a47abfe5cd650595c67a39c9c7Mechanism of the reaction of ebselen with endogenous thiols: dihydrolipoate is a better cofactor than glutathione in the peroxidase activity of ebselenHaenen, Guido R. M. M.; De Rooij, Ben M.; Vermeulen, Nico P. E.; Bast, AaltMolecular Pharmacology (1990), 37 (3), 412-22CODEN: MOPMA3; ISSN:0026-895X.The therapeutic effect of ebselen has been linked to its peroxidase activity. In the present study, the peroxidase activity of ebselen toward H2O2 with the endogenous thiols GSH and dihydrolipoate [L(SH)2] as cofactors was detd. When GSH was used, peroxide removal was described by a ter uni ping pong mechanism with Dalziel coeffs. for GSH and H2O2 of 0.165 and 0.081 mM min, resp. When L(SH)2 was used, peroxidase activity was independent of the concn. of L(SH)2, in the concn. range studied (5 μM to 2 mM) and peroxide removal was only dependent on the concn. of H2O2 and ebselen, with the second-order rate const. being 12.3 mM-1 min-1. To elucidate the difference between GSH and L(SH)2, the mol. mechanism of the peroxidase activity of ebselen was investigated, using UV spectrophotometry, HPLC, 77Se NMR, and mass spectrometry. GSH was found to react quickly with ebselen to give a selenenyl sulfide, an adduct of GSH to ebselen. Subsequently, the GSH-selenenyl sulfide is converted into the diselenide of ebselen. Finally the diselenide reacts with a peroxide and ebselen is regenerated. The formation by GSH of the diselenide from the GSH-selenenyl sulfide of ebselen is slow and linearly dependent on the concn. of free thiol; however, no net consumption of GSH was obsd. Furthermore, it is likely that a selenol is an intermediate in diselenide formation. After reaction between ebselen and L(SH)2 the diselenide of ebselen was immediately detected. The fast formation of the diselenide with L(SH)2 vs. the slow formation of the diselenide with GSH accounts for our observation that L(SH)2 is a better cofactor than GSH in the peroxidase activity of ebselen. These results suggest that the interaction between ebselen and L(SH)2 might be of major importance in the mechanism by which ebselen exerts its therapeutic effect.
- 28Wagner, G.; Schuch, G.; Akerboom, T. P.; Sies, H. Transport of ebselen in plasma and its transfer to binding sites in the hepatocyte. Biochem. Pharmacol. 1994, 48 (6), 1137– 1144, DOI: 10.1016/0006-2952(94)90150-328https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXmt1WjsL8%253D&md5=78392c5dbfaa4f9e7a5476146b852a55Transport of ebselen in plasma and its transfer to binding sites in the hepatocyteWagner, Gunter; Schuch, Gunter; Akerboom, Theodorus P. M.; Sies, HelmutBiochemical Pharmacology (1994), 48 (6), 1137-44CODEN: BCPCA6; ISSN:0006-2952.In vivo transport in plasma and in vitro transfer of ebselen to binding sits in the hepatocyte were studied. More than 90% of i.v. administered ebselen in mouse plasma is bound by selenium-sulfur bonds to reactive thiols in serum albumin. In in vitro expts. the uptake of [14C]-ebselen from a complex prepd. with bovine serum albumin (BSA) was detd. in isolated perfused rat liver. Radioactive ebselen metabolites were excreted into bile. In isolated hepatocytes, radioactivity was bound to all subcellular organelles. Ebselen is transferred from the BSA complex to membrane-assocd. proteins after reductive cleavage of the Se-S bond effected by endogenous protein thiols. In contrast, when proteins were sepd. by dialysis membranes, ebselen transfer from its BSA complex occurred only in the presence of externally added reductants. Among the physiol. reductants tested, ebselen release from the BSA complex was highest with glutathione (75%) and lowest with ascorbic acid (less than 10%). Quant. release of ebselen from its BSA complex was only achieved by the combined action of reductant, notably 2-mercaptoethanol, and guanidine thiocyanate, suggesting that ebselen interacts with proteins by covalent Se-S bonds as well as by ionic charge interactions.
- 29Singh, N.; Halliday, A. C.; Thomas, J. M.; Kuznetsova, O. V.; Baldwin, R.; Woon, E. C.; Aley, P. K.; Antoniadou, I.; Sharp, T.; Vasudevan, S. R.; Churchill, G. C. A safe lithium mimetic for bipolar disorder. Nat. Commun. 2013, 4, 1332, DOI: 10.1038/ncomms232029https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3s3nvVSqsw%253D%253D&md5=682e37608458f5d24a55405d07a0e5b4A safe lithium mimetic for bipolar disorderSingh Nisha; Halliday Amy C; Thomas Justyn M; Kuznetsova Olga V; Baldwin Rhiannon; Woon Esther C Y; Aley Parvinder K; Antoniadou Ivi; Sharp Trevor; Vasudevan Sridhar R; Churchill Grant CNature communications (2013), 4 (), 1332 ISSN:.Lithium is the most effective mood stabilizer for the treatment of bipolar disorder, but it is toxic at only twice the therapeutic dosage and has many undesirable side effects. It is likely that a small molecule could be found with lithium-like efficacy but without toxicity through target-based drug discovery; however, therapeutic target of lithium remains equivocal. Inositol monophosphatase is a possible target but no bioavailable inhibitors exist. Here we report that the antioxidant ebselen inhibits inositol monophosphatase and induces lithium-like effects on mouse behaviour, which are reversed with inositol, consistent with a mechanism involving inhibition of inositol recycling. Ebselen is part of the National Institutes of Health Clinical Collection, a chemical library of bioavailable drugs considered clinically safe but without proven use. Therefore, ebselen represents a lithium mimetic with the potential both to validate inositol monophosphatase inhibition as a treatment for bipolar disorder and to serve as a treatment itself.
- 30Johnson, T. W.; Dress, K. R.; Edwards, M. Using the Golden Triangle to optimize clearance and oral absorption. Bioorg. Med. Chem. Lett. 2009, 19 (19), 5560– 5564, DOI: 10.1016/j.bmcl.2009.08.04530https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFaktbfE&md5=3cfed1288ec451eb4419f1db753ae4b4Using the Golden Triangle to optimize clearance and oral absorptionJohnson, Ted W.; Dress, Klaus R.; Edwards, MartinBioorganic & Medicinal Chemistry Letters (2009), 19 (19), 5560-5564CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The Golden Triangle is a visualization tool developed from in vitro permeability, in vitro clearance and computational data designed to aid medicinal chemists in achieving metabolically stable, permeable and potent drug candidates. Classifying compds. as permeable and stable and plotting mol. wt. (MW) vs. octanol:buffer (pH 7.4) distribution coeffs. (log D) or estd. octanol:buffer (pH 7.4) distribution coeffs. (elog D) reveals useful trends. Anal. of at least 2 orthogonal trends, such as permeability and clearance, can be extremely effective in balancing and optimizing multiple properties. In addn., mol. wt. and log D impact potency-efficiency calcns., allowing potency, clearance and permeability to be optimized simultaneously.
- 31Johnson, T. W.; Gallego, R. A.; Edwards, M. P. Lipophilic Efficiency as an Important Metric in Drug Design. J. Med. Chem. 2018, 61 (15), 6401– 6420, DOI: 10.1021/acs.jmedchem.8b0007731https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXmsVahsbg%253D&md5=29f38880914fa75c33be3a640ace4309Lipophilic Efficiency as an Important Metric in Drug DesignJohnson, Ted W.; Gallego, Rebecca A.; Edwards, Martin P.Journal of Medicinal Chemistry (2018), 61 (15), 6401-6420CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Lipophilic efficiency (LipE) is an important metric that has been increasingly applied in drug discovery medicinal chem. lead optimization programs. In this perspective, using literature drug discovery examples, we discuss the concept of rigorously applying LipE to guide medicinal chem. lead optimization toward drug candidates with potential for superior in vivo efficacy and safety, esp. when guided by physiochem. property-based optimization (PPBO). Also highlighted are examples of small structural modifications such as addn. of single atoms, small functional groups, and cyclizations that produce large increases in LipE. Understanding the factors that may contribute to LipE changes through anal. of ligand-protein crystal structures and using structure-based drug design (SBDD) to increase LipE by design is also discussed. Herein we advocate for use of LipE anal. coupled with PPBO and SBDD as an efficient mechanism for drug design.
- 32Hopkins, A. L.; Keseru, G. M.; Leeson, P. D.; Rees, D. C.; Reynolds, C. H. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discovery 2014, 13 (2), 105– 121, DOI: 10.1038/nrd416332https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1SmtLk%253D&md5=344030e59f499180f79d311302a27532The role of ligand efficiency metrics in drug discoveryHopkins, Andrew L.; Keserue, Gyoergy M.; Leeson, Paul D.; Rees, David C.; Reynolds, Charles H.Nature Reviews Drug Discovery (2014), 13 (2), 105-121CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The judicious application of ligand or binding efficiency metrics, which quantify the mol. properties required to obtain binding affinity for a drug target, is gaining traction in the selection and optimization of fragments, hits and leads. Retrospective anal. of recently marketed oral drugs shows that they frequently have highly optimized ligand efficiency values for their targets. Optimizing ligand efficiency metrics based on both mol. mass and lipophilicity, when set in the context of the specific target, has the potential to ameliorate the inflation of these properties that has been obsd. in current medicinal chem. practice, and to increase the quality of drug candidates.
- 33Raymer, B.; Bhattacharya, S. K. Lead-like Drugs: A Perspective. J. Med. Chem. 2018, 61 (23), 10375– 10384, DOI: 10.1021/acs.jmedchem.8b0040733https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtl2is73O&md5=e9fc17d69c7d671459fdaf77d6721829Lead-like Drugs: A PerspectiveRaymer, Brian; Bhattacharya, Samit K.Journal of Medicinal Chemistry (2018), 61 (23), 10375-10384CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Lead-like drugs, or drugs below mol. wt. 300, are an important and sometimes overlooked component of the current pharmacopeia and contemporary medicinal chem. practice. To examine the recent state-of-the-art in lead-like drug discovery, we surveyed recent drug approvals from 2011 to 2017 and top 200 prescribed medications, as well as provide case studies on recently approved lead-like drugs. Many of these recent drugs are close analogs of previously known drugs or natural substrates, with a key focus of their medicinal chem. optimization being the choice of a low mol. wt. starting point and maintaining low mol. wt. during the optimization. However, the identification of low mol. wt. starting points may be limited by the availability of suitable low mol. wt. screening sets. To increase the discovery rate of lead-like drugs, we suggest an increased focus on inclusion and prosecution of lead-like starting points in screening libraries.
- 34Weiss, M. M.; Dineen, T. A.; Marx, I. E.; Altmann, S.; Boezio, A.; Bregman, H.; Chu-Moyer, M.; DiMauro, E. F.; Feric Bojic, E.; Foti, R. S.; Gao, H.; Graceffa, R.; Gunaydin, H.; Guzman-Perez, A.; Huang, H.; Huang, L.; Jarosh, M.; Kornecook, T.; Kreiman, C. R.; Ligutti, J.; La, D. S.; Lin, M. J.; Liu, D.; Moyer, B. D.; Nguyen, H. N.; Peterson, E. A.; Rose, P. E.; Taborn, K.; Youngblood, B. D.; Yu, V.; Fremeau, R. T., Jr. Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic Liabilities. J. Med. Chem. 2017, 60 (14), 5969– 5989, DOI: 10.1021/acs.jmedchem.6b0185134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXktFOmtbw%253D&md5=6333a6b936e48f9d18cf9df10ea12853Sulfonamides as Selective NaV1.7 Inhibitors: Optimizing Potency and Pharmacokinetics While Mitigating Metabolic LiabilitiesWeiss, Matthew M.; Dineen, Thomas A.; Marx, Isaac E.; Altmann, Steven; Boezio, Alessandro; Bregman, Howard; Chu-Moyer, Margaret; DiMauro, Erin F.; Feric Bojic, Elma; Foti, Robert S.; Gao, Hua; Graceffa, Russell; Gunaydin, Hakan; Guzman-Perez, Angel; Huang, Hongbing; Huang, Liyue; Jarosh, Michael; Kornecook, Thomas; Kreiman, Charles R.; Ligutti, Joseph; La, Daniel S.; Lin, Min-Hwa Jasmine; Liu, Dong; Moyer, Bryan D.; Nguyen, Hanh N.; Peterson, Emily A.; Rose, Paul E.; Taborn, Kristin; Youngblood, Beth D.; Yu, Violeta; Fremeau, Robert T.Journal of Medicinal Chemistry (2017), 60 (14), 5969-5989CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Several reports have recently emerged regarding the identification of heteroarylsulfonamides as NaV1.7 inhibitors that demonstrate high levels of selectivity over other NaV isoforms. The optimization of a series of internal NaV1.7 leads that address a no. of metabolic liabilities including bioactivation, PXR activation, as well as CYP3A4 induction and inhibition led to the identification of potent and selective inhibitors that demonstrated favorable pharmacokinetic profiles and were devoid of the aforementioned liabilities. The key to achieving this within a series prone to transporter-mediated clearance was the identification of a small range of optimal cLogD values and the discovery of subtle PXR SAR that was not lipophilicity dependent. This enabled the identification of compd. 20, which was advanced into a target engagement pharmacodynamic model where it exhibited robust reversal of histamine-induced scratching bouts in mice.
- 35Dittmar, M.; Lee, J. S.; Whig, K.; Segrist, E.; Li, M.; Kamalia, B.; Castellana, L.; Ayyanathan, K.; Cardenas-Diaz, F. L.; Morrisey, E. E.; Truitt, R.; Yang, W.; Jurado, K.; Samby, K.; Ramage, H.; Schultz, D. C.; Cherry, S. Drug repurposing screens reveal cell-type-specific entry pathways and FDA-approved drugs active against SARS-Cov-2. Cell Rep. 2021, 35 (1), 108959, DOI: 10.1016/j.celrep.2021.10895935https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnsVWju7w%253D&md5=52eaa65b358385380f995e46d3c7703dDrug repurposing screens reveal cell-type-specific entry pathways and FDA-approved drugs active against SARS-Cov-2Dittmar, Mark; Lee, Jae Seung; Whig, Kanupriya; Segrist, Elisha; Li, Minghua; Kamalia, Brinda; Castellana, Lauren; Ayyanathan, Kasirajan; Cardenas-Diaz, Fabian L.; Morrisey, Edward E.; Truitt, Rachel; Yang, Wenli; Jurado, Kellie; Samby, Kirandeep; Ramage, Holly; Schultz, David C.; Cherry, SaraCell Reports (2021), 35 (1), 108959CODEN: CREED8; ISSN:2211-1247. (Cell Press)There is an urgent need for antivirals to treat the newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To identify new candidates, we screen a repurposing library of ~ 3,000 drugs. Screening in Vero cells finds few antivirals, while screening in human Huh7.5 cells validates 23 diverse antiviral drugs. Extending our studies to lung epithelial cells, we find that there are major differences in drug sensitivity and entry pathways used by SARS-CoV-2 in these cells. Entry in lung epithelial Calu-3 cells is pH independent and requires TMPRSS2, while entry in Vero and Huh7.5 cells requires low pH and triggering by acid-dependent endosomal proteases. Moreover, we find nine drugs are antiviral in respiratory cells, seven of which have been used in humans, and three are US Food and Drug Administration (FDA) approved, including cyclosporine. We find that the antiviral activity of cyclosporine is targeting Cyclophilin rather than calcineurin, revealing essential host targets that have the potential for rapid clin. implementation.
- 36Jang, W. D.; Jeon, S.; Kim, S.; Lee, S. Y. Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assay. Proc. Natl. Acad. Sci. U. S. A. 2021, 118 (30), e2024302118 DOI: 10.1073/pnas.202430211836https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhs1KmsbjN&md5=d37de60099bc07ebf242e4a17132e153Drugs repurposed for COVID-19 by virtual screening of 6,218 drugs and cell-based assayJang, Woo Dae; Jeon, Sangeun; Kim, Seungtaek; Lee, Sang YupProceedings of the National Academy of Sciences of the United States of America (2021), 118 (30), e2024302118CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The COVID-19 pandemic caused by SARS-CoV-2 is an unprecedentedly significant health threat, prompting the need for rapidly developing antiviral drugs for the treatment. Drug repurposing is currently 1 of the most tangible options for rapidly developing drugs for emerging and reemerging viruses. In general, drug repurposing starts with virtual screening of approved drugs employing various computational methods. However, the actual hit rate of virtual screening is very low, and most of the predicted compds. are false positives. Here, we developed a strategy for virtual screening with much reduced false positives through incorporating predocking filtering based on shape similarity and postdocking filtering based on interaction similarity. We applied this advanced virtual screening approach to repurpose 6218 approved and clin. trial drugs for COVID-19. All 6218 compds. were screened against main protease and RNA-dependent RNA polymerase of SARS-CoV-2, resulting in 15 and 23 potential repurposed drugs, resp. Among them, 7 compds. can inhibit SARS-CoV-2 replication in Vero cells. Three of these drugs, emodin, omipalisib, and tipifarnib, show anti-SARS-CoV-2 activities in human lung cells, Calu-3. Notably, the activity of omipalisib is 200-fold higher than that of remdesivir in Calu-3. Furthermore, 3 drug combinations, omipalisib/remdesivir, tipifarnib/omipalisib, and tipifarnib/remdesivir, show strong synergistic effects in inhibiting SARS-CoV-2. Such drug combination therapy improves antiviral efficacy in SARS-CoV-2 infection and reduces the risk of each drug's toxicity. The drug repurposing strategy reported here will be useful for rapidly developing drugs for treating COVID-19 and other viruses.
- 37Choi, S. W.; Shin, J. S.; Park, S. J.; Jung, E.; Park, Y. G.; Lee, J.; Kim, S. J.; Park, H. J.; Lee, J. H.; Park, S. M.; Moon, S. H.; Ban, K.; Go, Y. Y. Antiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytes. Antiviral Res. 2020, 184, 104955, DOI: 10.1016/j.antiviral.2020.10495537https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitl2nsbfJ&md5=2a4723ae20790ada7df5ecabe730a2bdAntiviral activity and safety of remdesivir against SARS-CoV-2 infection in human pluripotent stem cell-derived cardiomyocytesChoi, Seong Woo; Shin, Jin Soo; Park, Soon-Jung; Jung, Eunhye; Park, Yun-Gwi; Lee, Jiho; Kim, Sung Joon; Park, Hun-Jun; Lee, Jung-Hoon; Park, Sung-Min; Moon, Sung-Hwan; Ban, Kiwon; Go, Yun YoungAntiviral Research (2020), 184 (), 104955CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is considered as the most significant global public health crisis of the century. Several drug candidates have been suggested as potential therapeutic options for COVID-19, including remdesivir, currently the only authorized drug for use under an Emergency Use Authorization. However, there is only limited information regarding the safety profiles of the proposed drugs, in particular drug-induced cardiotoxicity. Here, we evaluated the antiviral activity and cardiotoxicity of remdesivir using cardiomyocytes-derived from human pluripotent stem cells (hPSC-CMs) as an alternative source of human primary cardiomyocytes (CMs). In this study, remdesivir exhibited up to 60-fold higher antiviral activity in hPSC-CMs compared to Vero E6 cells; however, it also induced moderate cardiotoxicity in these cells. To gain further insight into the drug-induced arrhythmogenic risk, we assessed QT interval prolongation and automaticity of remdesivir-treated hPSC-CMs using a multielectrode array (MEA). As a result, the data indicated a potential risk of QT prolongation when remdesivir is used at concns. higher than the estd. peak plasma concn. Therefore, we conclude that close monitoring of the electrocardiog./QT interval should be advised in SARS-CoV-2-infected patients under remdesivir medication, in particular individuals with pre-existing heart conditions.
- 38Tiwari, S. K.; Wang, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Revealing Tissue-Specific SARS-CoV-2 Infection and Host Responses using Human Stem Cell-Derived Lung and Cerebral Organoids. Stem Cell Rep. 2021, 16 (3), 437– 445, DOI: 10.1016/j.stemcr.2021.02.00538https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXls1ersbw%253D&md5=e12410b74f51084672e12fab3eb5230eRevealing tissue-specific SARS-CoV-2 infection and host responses using human stem cell-derived lung and cerebral organoidsTiwari, Shashi Kant; Wang, Shaobo; Smith, Davey; Carlin, Aaron F.; Rana, Tariq M.Stem Cell Reports (2021), 16 (3), 437-445CODEN: SCRTFL; ISSN:2213-6711. (Cell Press)COVID-19 is a transmissible respiratory disease caused by a novel coronavirus, SARS-CoV-2, and has become a global health emergency. There is an urgent need for robust and practical in vitro model systems to investigate viral pathogenesis. Here, we generated human induced pluripotent stem cell (iPSC)-derived lung organoids (LORGs), cerebral organoids (CORGs), neural progenitor cells (NPCs), neurons, and astrocytes. LORGs contg. epithelial cells, alveolar types 1 and 2, highly express ACE2 and TMPRSS2 and are permissive to SARS-CoV-2 infection. SARS-CoV-2 infection induces interferons, cytokines, and chemokines and activates crit. inflammasome pathway genes. Spike protein inhibitor, EK1 peptide, and TMPRSS2 inhibitors (camostat/nafamostat) block viral entry in LORGs. Conversely, CORGs, NPCs, astrocytes, and neurons express low levels of ACE2 and TMPRSS2 and correspondingly are not highly permissive to SARS-CoV-2 infection. Infection in neuronal cells activates TLR3/7, OAS2, complement system, and apoptotic genes. These findings will aid in understanding COVID-19 pathogenesis and facilitate drug discovery.
- 39Li, N.; Hui, H.; Bray, B.; Gonzalez, G. M.; Zeller, M.; Anderson, K. G.; Knight, R.; Smith, D.; Wang, Y.; Carlin, A. F.; Rana, T. M. METTL3 regulates viral m6A RNA modification and host cell innate immune responses during SARS-CoV-2 infection. Cell Rep. 2021, 35 (6), 109091, DOI: 10.1016/j.celrep.2021.10909139https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhtVShu7zP&md5=938ee9a0e2de397723df26374d4e2612METTL3 regulates viral m6A RNA modification and host cell innate immune responses during SARS-CoV-2 infectionLi, Na; Hui, Hui; Bray, Bill; Gonzalez, Gwendolyn Michelle; Zeller, Mark; Anderson, Kristian G.; Knight, Rob; Smith, Davey; Wang, Yinsheng; Carlin, Aaron F.; Rana, Tariq M.Cell Reports (2021), 35 (6), 109091CODEN: CREED8; ISSN:2211-1247. (Cell Press)It is urgent and important to understand the relationship of the widespread severe acute respiratory syndrome coronavirus clade 2 (SARS-CoV-2) with host immune response and study the underlining mol. mechanism. N6-methylation of adenosine (m6A) in RNA regulates many physiol. and disease processes. Here, we investigate m6A modification of the SARS-CoV-2 gene in regulating the host cell innate immune response. Our data show that the SARS-CoV-2 virus has m6A modifications that are enriched in the 3' end of the viral genome. We find that depletion of the host cell m6A methyltransferase METTL3 decreases m6A levels in SARS-CoV-2 and host genes, and m6A redn. in viral RNA increases RIG-I binding and subsequently enhances the downstream innate immune signaling pathway and inflammatory gene expression. METTL3 expression is reduced and inflammatory genes are induced in patients with severe coronavirus disease 2019 (COVID-19). These findings will aid in the understanding of COVID-19 pathogenesis and the design of future studies regulating innate immunity for COVID-19 treatment.
- 40Wang, S.; Li, W.; Hui, H.; Tiwari, S. K.; Zhang, Q.; Croker, B. A.; Rawlings, S.; Smith, D.; Carlin, A. F.; Rana, T. M. Cholesterol 25-Hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterol. EMBO J. 2020, 39 (21), e106057 DOI: 10.15252/embj.202010605740https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFyisbvL&md5=7b37216366d03b329695f0703c5e96dfCholesterol 25-Hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterolWang, Shaobo; Li, Wanyu; Hui, Hui; Tiwari, Shashi Kant; Zhang, Qiong; Croker, Ben A.; Rawlings, Stephen; Smith, Davey; Carlin, Aaron F.; Rana, Tariq M.EMBO Journal (2020), 39 (21), e106057CODEN: EMJODG; ISSN:0261-4189. (Wiley-VCH Verlag GmbH & Co. KGaA)Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2 and has spread across the globe. SARS-CoV-2 is a highly infectious virus with no vaccine or antiviral therapy available to control the pandemic; therefore, it is crucial to understand the mechanisms of viral pathogenesis and the host immune responses to SARS-CoV-2. SARS-CoV-2 is a new member of the betacoronavirus genus like other closely related viruses including SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Both SARS-CoV and MERS-CoV have caused serious outbreaks and epidemics in the past 18 yr. We report that 1 of the interferon-stimulated genes (ISGs), cholesterol 25-hydroxylase (CH25H), is induced by SARS-CoV-2 infection in vitro and in COVID-19-infected patients. CH25H converts cholesterol to 25-hydroxycholesterol (25HC) and 25HC shows broad anti-coronavirus activity by blocking membrane fusion. Furthermore, 25HC inhibits USA-WA1/2020 SARS-CoV-2 infection in lung epithelial cells and viral entry in human lung organoids. Mechanistically, 25HC inhibits viral membrane fusion by activating the ER-localized acyl-CoA:cholesterol acyltransferase (ACAT) which leads to the depletion of accessible cholesterol from the plasma membrane. Altogether, our results shed light on a potentially broad antiviral mechanism by 25HC through depleting accessible cholesterol on the plasma membrane to suppress virus-cell fusion. Since 25HC is a natural product with no known toxicity at effective concns., it provides a potential therapeutic candidate for COVID-19 and emerging viral diseases in the future.
- 41Ma, C.; Hu, Y.; Townsend, J. A.; Lagarias, P. I.; Marty, M. T.; Kolocouris, A.; Wang, J. Ebselen, Disulfiram, Carmofur, PX-12, Tideglusib, and Shikonin Are Nonspecific Promiscuous SARS-CoV-2 Main Protease Inhibitors. ACS Pharmacol Transl Sci. 2020, 3 (6), 1265– 1277, DOI: 10.1021/acsptsci.0c0013041https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVajsrjK&md5=29d0e69db20f681ed4ef7cddd153cc6dEbselen, Disulfiram, Carmofur, PX-12, Tideglusib, and Shikonin Are Nonspecific Promiscuous SARS-CoV-2 Main Protease InhibitorsMa, Chunlong; Hu, Yanmei; Townsend, Julia Alma; Lagarias, Panagiotis I.; Marty, Michael Thomas; Kolocouris, Antonios; Wang, JunACS Pharmacology & Translational Science (2020), 3 (6), 1265-1277CODEN: APTSFN; ISSN:2575-9108. (American Chemical Society)Among the drug targets being investigated for SARS-CoV-2, the viral main protease (Mpro) is one of the most extensively studied. Mpro is a cysteine protease that hydrolyzes the viral polyprotein at more than 11 sites. It is highly conserved and has a unique substrate preference for glutamine in the P1 position. Therefore, Mpro inhibitors are expected to have broad-spectrum antiviral activity and a high selectivity index. Structurally diverse compds. have been reported as Mpro inhibitors. In this study, we investigated the mechanism of action of six previously reported Mpro inhibitors, ebselen, disulfiram, tideglusib, carmofur, shikonin, and PX-12, using a consortium of techniques including FRET-based enzymic assay, thermal shift assay, native mass spectrometry, cellular antiviral assays, and mol. dynamics simulations. Collectively, the results showed that the inhibition of Mpro by these six compds. is nonspecific and that the inhibition is abolished or greatly reduced with the addn. of reducing reagent 1,4-dithiothreitol (DTT). Without DTT, these six compds. inhibit not only Mpro but also a panel of viral cysteine proteases including SARS-CoV-2 papain-like protease and 2Apro and 3Cpro from enterovirus A71 (EV-A71) and EV-D68. However, none of the compds. inhibits the viral replication of EV-A71 or EV-D68, suggesting that the enzymic inhibition potency IC50 values obtained in the absence of DTT cannot be used to faithfully predict their cellular antiviral activity. Overall, we provide compelling evidence suggesting that these six compds. are nonspecific SARS-CoV-2 Mpro inhibitors and urge the scientific community to be stringent with hit validation.
- 42Weglarz-Tomczak, E.; Tomczak, J. M.; Talma, M.; Burda-Grabowska, M.; Giurg, M.; Brul, S. Identification of ebselen and its analogues as potent covalent inhibitors of papain-like protease from SARS-CoV-2. Sci. Rep. 2021, 11 (1), 3640, DOI: 10.1038/s41598-021-83229-642https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktVOntbY%253D&md5=bb905ef94cebd529c4fc53d196adc5a4Identification of ebselen and its analogues as potent covalent inhibitors of papain-like protease from SARS-CoV-2Weglarz-Tomczak, Ewelina; Tomczak, Jakub M.; Talma, Michal; Burda-Grabowska, Malgorzata; Giurg, Miroslaw; Brul, StanleyScientific Reports (2021), 11 (1), 3640CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)An efficient treatment against a COVID-19 disease, caused by the novel coronavirus SARS-CoV-2 (CoV2), remains a challenge. The papain-like protease (PLpro) from the human coronavirus is a protease that plays a crit. role in virus replication. Moreover, CoV2 uses this enzyme to modulate the host's immune system to its own benefit. Therefore, it represents a highly promising target for the development of antiviral drugs. We used Approx. Bayesian Computation tools, mol. modeling and enzyme activity studies to identify highly active inhibitors of the PLpro. We discovered organoselenium compds., ebselen and its structural analogs, as a novel approach for inhibiting the activity of PLproCoV2. Furthermore, we identified, for the first time, inhibitors of PLproCoV2 showing potency in the nanomolar range. Moreover, we found a difference between PLpro from SARS and CoV2 that can be correlated with the diverse dynamics of their replication, and, putatively to disease progression.
- 43Freitas, B. T.; Durie, I. A.; Murray, J.; Longo, J. E.; Miller, H. C.; Crich, D.; Hogan, R. J.; Tripp, R. A.; Pegan, S. D. Characterization and Noncovalent Inhibition of the Deubiquitinase and deISGylase Activity of SARS-CoV-2 Papain-Like Protease. ACS Infect. Dis. 2020, 6 (8), 2099– 2109, DOI: 10.1021/acsinfecdis.0c0016843https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpslylsbs%253D&md5=314ea59a92d60345f982e78e31e5b569Characterization and Noncovalent Inhibition of the Deubiquitinase and deISGylase Activity of SARS-CoV-2 Papain-Like ProteaseFreitas, Brendan T.; Durie, Ian A.; Murray, Jackelyn; Longo, Jaron E.; Miller, Holden C.; Crich, David; Hogan, Robert Jeff; Tripp, Ralph A.; Pegan, Scott D.ACS Infectious Diseases (2020), 6 (8), 2099-2109CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent for COVID-19, is a novel human betacoronavirus that is rapidly spreading worldwide. The outbreak currently includes over 3.7 million cases and 260,000 fatalities. As a betacoronavirus, SARS-CoV-2 encodes for a papain-like protease (PLpro) that is likely responsible for cleavage of the coronavirus (CoV) viral polypeptide. The PLpro is also responsible for suppression of host innate immune responses by virtue of its ability to reverse host ubiquitination and ISGylation events. Here, the biochem. activity of SARS-CoV-2 PLpro against ubiquitin (Ub) and interferon-stimulated gene product 15 (ISG15) substrates is evaluated, revealing that the protease has a marked redn. in its ability to process K48 linked Ub substrates compared to its counterpart in SARS-CoV. Addnl., its substrate activity more closely mirrors that of the PLpro from the Middle East respiratory syndrome coronavirus and prefers ISG15s from certain species including humans. Addnl., naphthalene based PLpro inhibitors are shown to be effective at halting SARS-CoV-2 PLpro activity as well as SARS-CoV-2 replication.
- 44Shin, D.; Mukherjee, R.; Grewe, D.; Bojkova, D.; Baek, K.; Bhattacharya, A.; Schulz, L.; Widera, M.; Mehdipour, A. R.; Tascher, G.; Geurink, P. P.; Wilhelm, A.; van der Heden van Noort, G. J.; Ovaa, H.; Muller, S.; Knobeloch, K. P.; Rajalingam, K.; Schulman, B. A.; Cinatl, J.; Hummer, G.; Ciesek, S.; Dikic, I. Papain-like protease regulates SARS-CoV-2 viral spread and innate immunity. Nature 2020, 587 (7835), 657– 662, DOI: 10.1038/s41586-020-2601-544https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVSqu7nP&md5=d6e55cd2e91c4a55f19abf2652973aabPapain-like protease regulates SARS-CoV-2 viral spread and innate immunityShin, Donghyuk; Mukherjee, Rukmini; Grewe, Diana; Bojkova, Denisa; Baek, Kheewoong; Bhattacharya, Anshu; Schulz, Laura; Widera, Marek; Mehdipour, Ahmad Reza; Tascher, Georg; Geurink, Paul P.; Wilhelm, Alexander; van der Heden van Noort, Gerbrand J.; Ovaa, Huib; Mueller, Stefan; Knobeloch, Klaus-Peter; Rajalingam, Krishnaraj; Schulman, Brenda A.; Cinatl, Jindrich; Hummer, Gerhard; Ciesek, Sandra; Dikic, IvanNature (London, United Kingdom) (2020), 587 (7835), 657-662CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The papain-like protease PLpro is an essential coronavirus enzyme that is required for processing viral polyproteins to generate a functional replicase complex and enable viral spread. PLpro is also implicated in cleaving proteinaceous post-translational modifications on host proteins as an evasion mechanism against host antiviral immune responses. We perform biochem., structural, and functional characterization of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PLpro (SCoV2-PLpro) and outline differences with SARS-CoV PLpro (SCoV-PLpro) in regulation of host interferon and NF-κB pathways. SCoV2-PLpro and SCoV-PLpro share 83% sequence identity but exhibit different host substrate preferences; SCoV2-PLpro preferentially cleaves the ubiquitin-like interferon-stimulated gene 15 protein (ISG15), whereas SCoV-PLpro predominantly targets ubiquitin chains. The crystal structure of SCoV2-PLpro in complex with ISG15 reveals distinctive interactions with the N-terminal ubiquitin-like domain of ISG15, highlighting the high affinity and specificity of these interactions. Furthermore, upon infection, SCoV2-PLpro contributes to the cleavage of ISG15 from interferon responsive factor 3 (IRF3) and attenuates type I interferon responses. Notably, inhibition of SCoV2-PLpro with GRL-0617 impairs the virus-induced cytopathogenic effect, maintains the antiviral interferon pathway and reduces viral replication in infected cells. These results highlight a potential dual therapeutic strategy in which targeting of SCoV2-PLpro can suppress SARS-CoV-2 infection and promote antiviral immunity.
- 45Klemm, T.; Ebert, G.; Calleja, D. J; Allison, C. C; Richardson, L. W; Bernardini, J. P; Lu, B. G.; Kuchel, N. W; Grohmann, C.; Shibata, Y.; Gan, Z. Y.; Cooney, J. P; Doerflinger, M.; Au, A. E; Blackmore, T. R; Heden van Noort, G. J; Geurink, P. P; Ovaa, H.; Newman, J.; Riboldi-Tunnicliffe, A.; Czabotar, P. E; Mitchell, J. P; Feltham, R.; Lechtenberg, B. C; Lowes, K. N; Dewson, G.; Pellegrini, M.; Lessene, G.; Komander, D. Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2. EMBO J. 2020, 39 (18), e106275 DOI: 10.15252/embj.202010627545https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1KgsrbE&md5=3d6a13fc99f05c4fdd6666e8fa72bbd7Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2Klemm, Theresa; Ebert, Gregor; Calleja, Dale J.; Allison, Cody C.; Richardson, Lachlan W.; Bernardini, Jonathan P.; Lu, Bernadine G. C.; Kuchel, Nathan W.; Grohmann, Christoph; Shibata, Yuri; Gan, Zhong Yan; Cooney, James P.; Doerflinger, Marcel; Au, Amanda E.; Blackmore, Timothy R.; van der Heden van Noort, Gerbrand J.; Geurink, Paul P.; Ovaa, Huib; Newman, Janet; Riboldi-Tunnicliffe, Alan; Czabotar, Peter E.; Mitchell, Jeffrey P.; Feltham, Rebecca; Lechtenberg, Bernhard C.; Lowes, Kym N.; Dewson, Grant; Pellegrini, Marc; Lessene, Guillaume; Komander, DavidEMBO Journal (2020), 39 (18), e106275CODEN: EMJODG; ISSN:0261-4189. (Wiley-VCH Verlag GmbH & Co. KGaA)The SARS-CoV-2 coronavirus encodes an essential papain-like protease domain as part of its non-structural protein (nsp)-3, namely SARS2 PLpro, that cleaves the viral polyprotein, but also removes ubiquitin-like ISG15 protein modifications as well as, with lower activity, Lys48-linked polyubiquitin. Structures of PLpro bound to ubiquitin and ISG15 reveal that the S1 ubiquitin-binding site is responsible for high ISG15 activity, while the S2 binding site provides Lys48 chain specificity and cleavage efficiency. To identify PLpro inhibitors in a repurposing approach, screening of 3,727 unique approved drugs and clin. compds. against SARS2 PLpro identified no compds. that inhibited PLpro consistently or that could be validated in counterscreens. More promisingly, non-covalent small mol. SARS PLpro inhibitors also target SARS2 PLpro, prevent self-processing of nsp3 in cells and display high potency and excellent antiviral activity in a SARS-CoV-2 infection model.
- 46Liu, G.; Lee, J.-H.; Parker, Z. M.; Acharya, D.; Chiang, J. J.; van Gent, M.; Riedl, W.; Davis-Gardner, M. E.; Wies, E.; Chiang, C.; Gack, M. U. ISG15-dependent activation of the sensor MDA5 is antagonized by the SARS-CoV-2 papain-like protease to evade host innate immunity. Nature Microbiology 2021, 6, 467– 478, DOI: 10.1038/s41564-021-00884-146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXms1Onsbs%253D&md5=a74cf2f655bfac4c2e8516b0f9f0f4e0ISG15-dependent activation of the sensor MDA5 is antagonized by the SARS-CoV-2 papain-like protease to evade host innate immunityLiu, GuanQun; Lee, Jung-Hyun; Parker, Zachary M.; Acharya, Dhiraj; Chiang, Jessica J.; van Gent, Michiel; Riedl, William; Davis-Gardner, Meredith E.; Wies, Effi; Chiang, Cindy; Gack, Michaela U.Nature Microbiology (2021), 6 (4), 467-478CODEN: NMAICH; ISSN:2058-5276. (Nature Research)Abstr.: Activation of the RIG-I-like receptors, retinoic-acid inducible gene I (RIG-I) and melanoma differentiation-assocd. protein 5 (MDA5), establishes an antiviral state by upregulating interferon (IFN)-stimulated genes (ISGs). Among these is ISG15, the mechanistic roles of which in innate immunity still remain enigmatic. In the present study, we report that ISG15 conjugation is essential for antiviral IFN responses mediated by the viral RNA sensor MDA5. ISGylation of the caspase activation and recruitment domains of MDA5 promotes its oligomerization and thereby triggers activation of innate immunity against a range of viruses, including coronaviruses, flaviviruses and picornaviruses. The ISG15-dependent activation of MDA5 is antagonized through direct de-ISGylation mediated by the papain-like protease of SARS-CoV-2, a recently emerged coronavirus that has caused the COVID-19 pandemic. Our work demonstrates a crucial role for ISG15 in the MDA5-mediated antiviral response, and also identifies a key immune evasion mechanism of SARS-CoV-2, which may be targeted for the development of new antivirals and vaccines to combat COVID-19.
- 47Jacobson, M. P.; Friesner, R. A.; Xiang, Z.; Honig, B. On the role of the crystal environment in determining protein side-chain conformations. J. Mol. Biol. 2002, 320 (3), 597– 608, DOI: 10.1016/S0022-2836(02)00470-947https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XltVKmu70%253D&md5=006de6bd2d0f233ab32d6798dc1a3fbcOn the Role of the Crystal Environment in Determining Protein Side-chain ConformationsJacobson, Matthew P.; Friesner, Richard A.; Xiang, Zhexin; Honig, BarryJournal of Molecular Biology (2002), 320 (3), 597-608CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Science Ltd.)The role of crystal packing in detg. the obsd. conformations of amino acid side-chains in protein crystals is investigated by (1) anal. of a database of proteins that have been crystd. in different unit cells (space group or unit cell dimensions) and (2) theor. predictions of side-chain conformations with the crystal environment explicitly represented. Both of these approaches indicate that the crystal environment plays an important role in detg. the conformations of polar side-chains on the surfaces of proteins. Inclusion of the crystal environment permits a more sensitive measurement of the achievable accuracy of side-chain prediction programs, when validating against structures obtained by x-ray crystallog. Our side-chain prediction program uses an all-atom force field and a Generalized Born model of solvation and is thus capable of modeling simple packing effects (i.e. van der Waals interactions), electrostatic effects, and desolvation, which are all important mechanisms by which the crystal environment impacts obsd. side-chain conformations. Our results are also relevant to the understanding of changes in side-chain conformation that may result from ligand docking and protein-protein assocn., insofar as the results reveal how side-chain conformations change in response to their local environment.
- 48Jacobson, M. P.; Pincus, D. L.; Rapp, C. S.; Day, T. J.; Honig, B.; Shaw, D. E.; Friesner, R. A. A hierarchical approach to all-atom protein loop prediction. Proteins: Struct., Funct., Genet. 2004, 55 (2), 351– 367, DOI: 10.1002/prot.1061348https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjtFKhsrc%253D&md5=e0eff655eeefb30ea00ae041ea9099c8A hierarchical approach to all-atom protein loop predictionJacobson, Matthew P.; Pincus, David L.; Rapp, Chaya S.; Day, Tyler J. F.; Honig, Barry; Shaw, David E.; Friesner, Richard A.Proteins: Structure, Function, and Bioinformatics (2004), 55 (2), 351-367CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The application of all-atom force fields (and explicit or implicit solvent models) to protein homol.-modeling tasks such as side-chain and loop prediction remains challenging both because of the expense of the individual energy calcns. and because of the difficulty of sampling the rugged all-atom energy surface. Here the authors address this challenge for the problem of loop prediction through the development of numerous new algorithms, with an emphasis on multiscale and hierarchical techniques. As a first step in evaluating the performance of the authors' loop prediction algorithm, the authors have applied it to the problem of reconstructing loops in native structures; the authors also explicitly include crystal packing to provide a fair comparison with crystal structures. In brief, large nos. of loops are generated by using a dihedral angle-based buildup procedure followed by iterative cycles of clustering, side-chain optimization, and complete energy minimization of selected loop structures. The authors evaluate this method by the largest test set yet used for validation of a loop prediction method, with a total of 833 loops ranging from 4 to 12 residues in length. Av./median backbone root-mean-square deviations (RMSDs) to the native structures (superimposing the body of the protein, not the loop itself) are 0.42/0.24 Å for 5 residue loops, 1.00/0.44 Å for 8 residue loops, and 2.47/1.83 Å for 11 residue loops. Median RMSDs are substantially lower than the avs. because of a small no. of outliers; the causes of these failures are examd. in some detail, and many can be attributed to errors in assignment of protonation states of titratable residues, omission of ligands from the simulation, and, in a few cases, probable errors in the exptl. detd. structures. When these obvious problems in the data sets are filtered out, av. RMSDs to the native structures improve to 0.43 Å for 5 residue loops, 0.84 Å for 8 residue loops, and 1.63 Å for 11 residue loops. In the vast majority of cases, the method locates energy min. that are lower than or equal to that of the minimized native loop, thus indicating that sampling rarely limits prediction accuracy. The overall results are, to the authors' knowledge, the best reported to date, and the authors attribute this success to the combination of an accurate all-atom energy function, efficient methods for loop buildup and side-chain optimization, and, esp. for the longer loops, the hierarchical refinement protocol.
- 49Donovan, S. F.; Pescatore, M. C. Method for measuring the logarithm of the octanol-water partition coefficient by using short octadecyl-poly(vinyl alcohol) high-performance liquid chromatography columns. J. Chromatogr A 2002, 952 (1–2), 47– 61, DOI: 10.1016/S0021-9673(02)00064-X49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XisV2gs7s%253D&md5=4c4aa02a23c1a496d3a9107d43329629Method for measuring the logarithm of the octanol-water partition coefficient by using short octadecyl-poly(vinyl alcohol) high-performance liquid chromatography columnsDonovan, Stephen F.; Pescatore, Mark C.Journal of Chromatography A (2002), 952 (1-2), 47-61CODEN: JCRAEY; ISSN:0021-9673. (Elsevier Science B.V.)A simple, quick, versatile and inexpensive HPLC method to est. the logarithm of the octanol-water partition coeff. (log Pow) employing a methanol-water gradient and a short octadecyl-poly(vinyl alc.) (ODP) column is described. This method is different from published HPLC-based log Pow methods because it uses retention times from a rapid methanol-water gradient to directly generate log Pow ests., rather than from a series of isocratic mixts. extrapolated to 100% water. These HPLC log Pow values have good precision and correlate well with traditional shake-flask log Pow values. If necessary, the log Pow detn. (including replications) can easily be carried out using only a milligram of sample. By suppressing ionization of acids and bases by the use of a buffer in the aq. phase, the method can measure the log Pow of neutral org. mols. at any pH between 2 and 13. The method can be used with impure material and is rapid, 7 min per run and 4 min equilibration; it lends itself to and has been utilized for high-throughput hydrophobicity detns. (we have now carried out thousands of HPLC log Pow measurements by this method).
- 50Box, K.; Bevan, C.; Comer, J.; Hill, A.; Allen, R.; Reynolds, D. High-throughput measurement of pKa values in a mixed-buffer linear pH gradient system. Anal. Chem. 2003, 75 (4), 883– 892, DOI: 10.1021/ac020329y50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXjt1yksg%253D%253D&md5=4050d66a7c92550c1efd346e76c392d9High-Throughput Measurement of pKa Values in a Mixed-Buffer Linear pH Gradient SystemBox, Karl; Bevan, Christopher; Comer, John; Hill, Alan; Allen, Ruth; Reynolds, DerekAnalytical Chemistry (2003), 75 (4), 883-892CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A procedure is described for measuring pKa values in a short time, e.g., 4 min/assay. Samples, as 10 mM solns., are prepd. in DMSO in 96-well plates. A flowing pH gradient is produced by mixing two buffer solns. contg. mixts. of weak acids and bases that do not absorb significantly in the UV above 250 nm. The sample soln. is dild. with water and then injected directly into the flowing gradient, which then passes through a diode array spectrophotometer measuring in the UV wavelength range. The buffer has been formulated so that its acid-base titrn. curve is linear over a wide pH range, such that the pH of the gradient is a linear function of time. The soln. pH in the measurement flow cell is therefore proportional to the time elapsed since the start of gradient generation. The sample's pKa values are calcd. from the change in UV absorbance at multiple wavelengths as a function of pH. The pKa values of 71 drugs have been measured, and results compare well with values measured by pH-metric or traditional UV methods. Rules are suggested for the rapid inspection of data and the choice of method for the calcn. of pKa from the data.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c00566.
Molecular formula strings(CSV)
Expanded synthetic methods and spectra for E01–30, structures and reported activities for known SARS-CoV-2 main protease inhibitors, docking poses, and individual antiviral dose–response curves for select compounds
(PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.