Synthetic Peptides That Antagonize the Angiotensin-Converting Enzyme-2 (ACE-2) Interaction with SARS-CoV-2 Receptor Binding Spike ProteinClick to copy article linkArticle link copied!
- Afsaneh SadremomtazAfsaneh SadremomtazXB20 Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9700 AD Groningen, The NetherlandsMore by Afsaneh Sadremomtaz
- Zayana M. Al-DahmaniZayana M. Al-DahmaniXB20 Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9700 AD Groningen, The NetherlandsDepartment of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The NetherlandsMore by Zayana M. Al-Dahmani
- Angel J. Ruiz-MorenoAngel J. Ruiz-MorenoXB20 Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9700 AD Groningen, The NetherlandsDepartamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Ciudad de Mexico 04510, MexicoUnidad Periférica de Investigación en Biomedicina Translacional, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Félix Cuevas 540, Ciudad de Mexico 03229, MexicoDoctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico 04510, MexicoMore by Angel J. Ruiz-Moreno
- Alessandra MontiAlessandra MontiInstitute of Biostructures and Bioimaging (IBB)-CNR, Via Mezzocannone, 16, 80134 Napoli, ItalyMore by Alessandra Monti
- Chao WangChao WangXB20 Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9700 AD Groningen, The NetherlandsMore by Chao Wang
- Taha AzadTaha AzadCenter for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, K1H 8L6 ON, CanadaDepartment of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, K1H 8M5 ON, CanadaMore by Taha Azad
- John C. BellJohn C. BellCenter for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, K1H 8L6 ON, CanadaDepartment of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, K1H 8M5 ON, CanadaMore by John C. Bell
- Nunzianna DotiNunzianna DotiInstitute of Biostructures and Bioimaging (IBB)-CNR, Via Mezzocannone, 16, 80134 Napoli, ItalyMore by Nunzianna Doti
- Marco A. Velasco-VelázquezMarco A. Velasco-VelázquezDepartamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Ciudad de Mexico 04510, MexicoUnidad Periférica de Investigación en Biomedicina Translacional, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Félix Cuevas 540, Ciudad de Mexico 03229, MexicoDoctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico 04510, MexicoMore by Marco A. Velasco-Velázquez
- Debora de JongDebora de JongDepartment of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The NetherlandsMore by Debora de Jong
- Jørgen de JongeJørgen de JongeCentre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3720BA Bilthoven, The NetherlandsMore by Jørgen de Jonge
- Jolanda SmitJolanda SmitDepartment of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The NetherlandsMore by Jolanda Smit
- Alexander DömlingAlexander DömlingXB20 Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9700 AD Groningen, The NetherlandsMore by Alexander Dömling
- Harry van Goor*Harry van Goor*Email: [email protected]Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9700RB Groningen, The NetherlandsMore by Harry van Goor
- Matthew R. Groves*Matthew R. Groves*Email: [email protected]XB20 Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9700 AD Groningen, The NetherlandsMore by Matthew R. Groves
Abstract
The SARS-CoV-2 viral spike protein S receptor-binding domain (S-RBD) binds ACE2 on host cells to initiate molecular events, resulting in intracellular release of the viral genome. Therefore, antagonists of this interaction could allow a modality for therapeutic intervention. Peptides can inhibit the S-RBD:ACE2 interaction by interacting with the protein–protein interface. In this study, protein contact atlas data and molecular dynamics simulations were used to locate interaction hotspots on the secondary structure elements α1, α2, α3, β3, and β4 of ACE2. We designed a library of discontinuous peptides based upon a combination of the hotspot interactions, which were synthesized and screened in a bioluminescence-based assay. The peptides demonstrated high efficacy in antagonizing the SARS-CoV-2 S-RBD:ACE2 interaction and were validated by microscale thermophoresis which demonstrated strong binding affinity (∼10 nM) of these peptides to S-RBD. We anticipate that such discontinuous peptides may hold the potential for an efficient therapeutic treatment for COVID-19.
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 and Discussion
Molecular Docking and Computational Modeling of ACE2:S-RBD Antagonistic Peptides
m/z (monoisotopic) | |||||
---|---|---|---|---|---|
entry | sequence | theor | exptl | Kd (nM) | IC50 (nM) |
peptide 1 | H-IEEQAKTFLDKFQHEVEEIYWQS-NH2 | 2895.397 | 2895.477 | 106 ± 1 | 11 ± 1 |
peptide 2 | H-QDKHEEDYQMYNKGDKED-NH2 | 2269.944 | 2270.011 | 102 ± 6 | 18 ± 2 |
peptide 3 | H -DKFNHEAEDLFYQSSLASWNYNT-NH2 | 2777.225 | 2777.304 | 245 ± 3 | 6 ± 4 |
peptide 4 | H-IDENARSYIDKFQHDAEEMWYQ-NH2 | 2786.228 | 2786.308 | 541 ± 5 | 32 ± 2 |
peptide 5 | H-IYALLENAEDYNLVN-NH2 | 1751.862 | 1751.920 | 13 ± 1 | 9 ± 4 |
peptide 6 | H-SRDKHEEHEKENDRGQ-NH2 | 1991.905 | 1991.966 | 46 ± 5 | 10 ± 5 |
Theoretical and experimental molecular weight, half-maximal inhibitory concentration (IC50) using a luciferase assay and binding affinities of SARS-CoV-2:ACE2-antagonist peptides (determined using) MST are also shown. Peptides were synthesized on solid phase using the F-moc strategy, have a free N-terminus, and are amidated at the C-terminus.
Figure 1
Figure 1. Interaction of ACE2 with S-RBD. (a) Surface representation of the complex between the receptor binding (S-RBD) domain of SARS-CoV-2 Spike protein (yellow) and the human ACE2 receptor (pink) (PDB ID: 6M0J). The portion of the ACE2 domain including main interacting residues of helices α1 (I21, Q24, T27, F28, D30, K31, H34, E35, E37, D38, Y41, Q42), α2 (L79, M82, Y83), and α3 (N330, K419, D430, E431) and β sheets β3 and β4 (K353, G354, D355, and R357) are drawn in green. (b) A closer view displays the interacting residues at the interface site. Figure created by PyMol (Molecular Graphics System, ver. 1.2r3pre, Schrödinger, LLC).
Figure 2
Figure 2. Stick representation of residues involved in the interprotomer interaction of S-RBD. (a) Side view of the surface representation of the interactions within ACE2 and S-RRBD (PDB ID: 6M0J). (b) Residues involved in the subunit interaction are shown in green (cartoon transparency is set to 40%). Four contact regions are located in the α1, α2and α3 helices and in β3 and β4 of ACE2 and S-RBD. Figure created by PyMol (Molecular Graphics System, ver. 1.2r3pre, Schrödinger, LLC).
entry | electrostatic energy score (arbitrary units of energy) | van der Waals energy score (arbitrary units of energy) | score | buried surface area (A•2) | effect |
---|---|---|---|---|---|
peptide 1 | –284.989 ± 2.3 | –113.23 ± 1.1 | –94.364 ± 7.2 | 927.565 ± 32 | B |
peptide 2 | –386.163 ± 1.7 | –127.31 ± 2.2 | –104.789 ± 4.3 | 1024.23 ± 21 | C |
peptide 3 | –272.940 ± 2.4 | –111.43 ± 0.17 | –93.334 ± 9.6 | 995.967 ± 34 | B |
peptide 4 | –265.347 ± 1.1 | –105.61 ± 0.9 | –87.56 ± 3.5 | 1021.25 ± 29 | B |
peptide 5 | –388.163 ± 4.2 | –190.20 ± 1.4 | –155.53 ± 9.2 | 1123.57 ± 37 | D |
peptide 6 | –383.163 ± 3.3 | –187.36 ± 1.9 | –143.034 ± 10.13 | 1017.77 ± 41 | D |
Effect; The experimentally determined effect on interaction of ACE2 with S-RBD. A: No effect on interaction with S-RBD. B: Slightly inhibits interaction with S-RBD. C: Strongly inhibits interaction with S-RBD. D: Abolishes interaction with S-RBD.
Figure 3
Figure 3. (a) Heat maps representing the pairwise backbone RMSD matrix of SARS-CoV-2 S-RBD protein calculated for the backbone along 50 ns of MD simulation from systems including peptides with stable binding to S-RBD. The unliganded protein (apo-RBD) is included for comparison. The simulation corresponding to apo-RBD displays a higher RMSD in comparison with the matrices originated for peptides 1, 2, 5, and 6. Indicating a less flexible conformation of S-RBD. (b) The unliganded protein (ACE2:S-RBD) is included for comparison
Figure 4
Figure 4. α-Carbon RMSF analysis for the peptide-S-RBD systems. (a) α-Carbon RMSF profiles of all studied peptides, apo-S-RBD, and ACE2 are presented for comparison. (b) RMSF structural representation of apo-S-RBD and ACE2:S-RBD. (c) structural representation of peptide:S-RBD complexes including the binding of the peptides across 10 representative snapshots. Normalized scale for peptides 1–5; peptide 6 is presented with its own scale.
Biophysical Characterization of Peptides of ACE2-Antagonist Peptides
Synthetic Peptides Efficiently Block the Interaction of ACE2 with S-RBD
Figure 5
Figure 5. (a) Binding analysis for the interaction between S-RBD and ACE2. (a) Luciferase-based assay. 293T cells were transfected with the ACE2 or S-RBD expression constructs. 48 h post-transfection, and luciferase assays were performed on 20 μg total protein from cell lysates using FMZ as a substrate (n = 3, mean ± SD; one-way ANOVA, ***p < 0.005 relative to smBiT-ACE2 alone, Dunnett’s correction for multiple comparisons). (b) MST analysis of peptide 1–6 binding to recombinant S-RBD. The concentration of S-RBD is kept constant at 50 nM, while the ligand concentration varies from 12.5 μM to 0.19 nM. Serial titrations result in measurable changes in the fluorescence signal within a temperature gradient that can be used to calculate the dissociation constant (Kd). The curve is shown as ΔFnorm (change of Fnorm with respect to the zero-ligand concentration) against S-RBD concentration on a log scale.
Synthetic Peptides Bind to Purified S-RBD with Nanomolar Affinity
Conclusion
Experimental Section
Protein-Peptide Docking
Molecular Dynamics
Free Energy Calculations
Peptide Synthesis and Characterization
Determination of Peptide Concentrations
Intrinsic Fluorescence Analysis
Circular Dichroism (CD) Measurements
Size-Exclusion Chromatography Experiments
Plasmid Construction
Cell Culture
In Vitro NanoLuc Assay
Microscale Thermophoresis (MST)
Statistical Analysis
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c00477.
Additional figures illustrating molecular interaction from MD analysis of designed peptides and S-RBD, binding free energy of ACE2 derived peptides in complexes of S-RBD (binding energies of ACE2:S-RBD included for comparison), peptide LC-MS characterization, details of changes in hot spot area plot caused by aggregation of peptides, variation of fluorescence peak intensity with increasing concentration of peptides, CD spectra of peptides, overlay of emission spectra of samples dissolved in PBS and PBST, 1% (w/v) PEG8000 at 25 μM in 2.5% DMSO, and SEC of peptides; additional tables detailing key interactions between S-RBD and ACE2 identified using PiPreD, peptide sequences including replacement of some residues for improved targeting of S-RBD, analysis of data collected for the calculation of concentration of peptides in PBS and PBST, 1% (w/v) PEG8000 by UV spectroscopy analysis, theoretical, apparent molecular weight, and Rt values of ACE2-antagonist peptides as determined by SEC analysis, and dose–response results of the antiviral activity of peptides in a SARS-CoV-2 infection inhibition assay performed in VERO-E6 cells (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
This research was supported by private funding by the University of Groningen and University Medical Center Groningen. Support from the project PRIN2017 Prot. 2017M8R7N9 is gratefully acknowledged. Computational studies have implemented by a grant from Universidad Nacional Autónoma de México (UNAM) supercomputer “Miztli” (LANCAD-UNAM-DGTIC-386 2020-2021) and Ph.D. scholarship of A.J.R-M. as a doctoral student from Programa de Doctorado en Ciencias Biomédicas, UNAM with fellowship 584534 from CONACYT.
ACE2 | angiotensin-converting Enzyme 2 |
ANOVA | Analysis of variance |
CD | circular dichroism |
CH3CN | acetonitrile |
CTZ | coelenterazine |
DMF | dimethylformamide |
DIC | N,N′-diisopropylcarbodiimide |
DIPEA | diisopropylethylamine |
ESI-TOF-MS | electrospray ionization time-of-flight mass spectrometry |
FMZ | furimazine |
ITC | isothermal titration Calorimetry |
IC50 | half-maximal inhibitory concentration |
LC/MS | liquid chromatography–mass spectrometry |
mRNA | messenger RNA |
MD | molecular dynamics |
PPI | protein–protein interaction |
PBS | phosphate-buffered saline |
PEG | polyethylene glycol |
PBC | periodic boundary conditions |
PDA | photodiode array detector |
RNA | ribonucleic acid |
RMSD | root-mean-square deviation |
RMSF | root-mean-square fluctuation |
RLU | relative luminescence unit |
SARS-CoV-2 | severe acute respiratory syndrome coronavirus 2 |
S-RBD | spike-receptor binding domain |
SPR | surface plasmon resonance |
SD | standard deviation |
TIS | tri-isopropylsilane |
TFA | trifluoroacetic acid |
TOF | time-of-flight |
VEGF | vascular endothelial growth factor |
VEGFR | vascular endothelial growth factor receptor |
WHO | World Health Organization |
References
This article references 77 other publications.
- 1Liu, C. H.; Lu, C. H.; Wong, S. H.; Lin, L. T. Update on antiviral strategies against COVID-19: unmet needs and prospects. Front. Immunol. 2021, 11, 616595, DOI: 10.3389/fimmu.2020.616595Google ScholarThere is no corresponding record for this reference.
- 2Galindez, G.; Matschinske, J.; Rose, T. D.; Sadegh, S.; Salgado-Albarrán, M.; Späth, J.; Baumbach, J.; Pauling, J. K. Lessons from the COVID-19 pandemic for advancing computational drug repurposing strategies. Nat. Comput. Sci. 2021, 1, 33– 41, DOI: 10.1038/s43588-020-00007-6Google ScholarThere is no corresponding record for this reference.
- 3Hufsky, F.; Lamkiewicz, K.; Almeida, A.; Aouacheria, A.; Arighi, C. Computational strategies to combat COVID-19: useful tools to accelerate SARS-CoV-2 and coronavirus research. Briefings Bioinf. 2021, 22, 642– 663, DOI: 10.1093/bib/bbaa232Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFWht73E&md5=b79ea64cf7ce9c512e9481ea30416bc8Computational strategies to combat COVID-19: useful tools to accelerate SARS-CoV-2 and coronavirus researchHufsky, Franziska; Lamkiewicz, Kevin; Almeida, Alexandre; Aouacheria, Abdel; Arighi, Cecilia; Bateman, Alex; Baumbach, Jan; Beerenwinkel, Niko; Brandt, Christian; Cacciabue, Marco; Chuguransky, Sara; Drechsel, Oliver; Finn, Robert D.; Fritz, Adrian; Fuchs, Stephan; Hattab, Georges; Hauschild, Anne-Christin; Heider, Dominik; Hoffmann, Marie; Holzer, Martin; Hoops, Stefan; Kaderali, Lars; Kalvari, Ioanna; von Kleist, Max; Kmiecinski, Reno; Kuhnert, Denise; Lasso, Gorka; Libin, Pieter; List, Markus; Lochel, Hannah F.; Martin, Maria J.; Martin, Roman; Matschinske, Julian; McHardy, Alice C.; Mendes, Pedro; Mistry, Jaina; Navratil, Vincent; Nawrocki, Eric P.; O'Toole, Aine Niamh; Ontiveros-Palacios, Nancy; Petrov, Anton I.; Rangel-Pineros, Guillermo; Redaschi, Nicole; Reimering, Susanne; Reinert, Knut; Reyes, Alejandro; Richardson, Lorna; Robertson, David L.; Sadegh, Sepideh; Singer, Joshua B.; Theys, Kristof; Upton, Chris; Welzel, Marius; Williams, Lowri; Marz, ManjaBriefings in Bioinformatics (2021), 22 (2), 642-663CODEN: BBIMFX; ISSN:1477-4054. (Oxford University Press)A review. SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is a novel virus of the family Coronaviridae. The virus causes the infectious disease COVID-19. The biol. of coronaviruses has been studied for many years. However, bioinformatics tools designed explicitly for SARS-CoV-2 have only recently been developed as a rapid reaction to the need for fast detection, understanding and treatment of COVID-19. To control the ongoing COVID-19 pandemic, it is of utmost importance to get insight into the evolution and pathogenesis of the virus. In this review, we cover bioinformatics workflows and tools for the routine detection of SARS-CoV-2 infection, the reliable anal. of sequencing data, the tracking of the COVID-19 pandemic and evaluation of containment measures, the study of coronavirus evolution, the discovery of potential drug targets and development of therapeutic strategies. For each tool, we briefly describe its use case and how it advances research specifically for SARS-CoV-2. All tools are free to use and available online, either through web applications or public code repositories.
- 4V’kovski, P.; Kratzel, A.; Steiner, S.; Stalder, H.; Thiel, V. Coronavirus biology and replication: implications for SARS-CoV-2. Nat. Rev. Microbiol. 2021, 19, 155– 170, DOI: 10.1038/s41579-020-00468-6Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1GjsLjF&md5=14ddf54f3e569fe39eb4f3295cf603e6Coronavirus biology and replication: implications for SARS-CoV-2V'kovski, Philip; Kratzel, Annika; Steiner, Silvio; Stalder, Hanspeter; Thiel, VolkerNature Reviews Microbiology (2021), 19 (3), 155-170CODEN: NRMACK; ISSN:1740-1526. (Nature Research)A review. Abstr.: The SARS-CoV-2 pandemic and its unprecedented global societal and economic disruptive impact has marked the third zoonotic introduction of a highly pathogenic coronavirus into the human population. Although the previous coronavirus SARS-CoV and MERS-CoV epidemics raised awareness of the need for clin. available therapeutic or preventive interventions, to date, no treatments with proven efficacy are available. The development of effective intervention strategies relies on the knowledge of mol. and cellular mechanisms of coronavirus infections, which highlights the significance of studying virus-host interactions at the mol. level to identify targets for antiviral intervention and to elucidate crit. viral and host determinants that are decisive for the development of severe disease. In this Review, we summarize the first discoveries that shape our current understanding of SARS-CoV-2 infection throughout the intracellular viral life cycle and relate that to our knowledge of coronavirus biol. The elucidation of similarities and differences between SARS-CoV-2 and other coronaviruses will support future preparedness and strategies to combat coronavirus infections.
- 5Zhang, 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 α-ketoamide inhibitors. Science 2020, 368, 409– 412, DOI: 10.1126/science.abb3405Google Scholar5https://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.
- 6Jin, 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 Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289– 293, DOI: 10.1038/s41586-020-2223-yGoogle Scholar6https://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.
- 7Hussain, S.; Xie, Y. J.; Li, D.; Malik, S. I.; Hou, J. C.; Leung, E. L.; Fan, X. X. Current strategies against COVID-19. Chin. Med. 2020, 15, 70, DOI: 10.1186/s13020-020-00353-7Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlOhurzJ&md5=a8d06ec5240ed4d5224dfb3be7c76635Current strategies against COVID-19Hussain, Shahid; Xie, Ya-Jia; Li, Dan; Malik, Shaukat Iqbal; Hou, Jin-cai; Leung, Elaine Lai-Han; Fan, Xing-XingChinese Medicine (London, United Kingdom) (2020), 15 (1), 70CODEN: CMHEBK; ISSN:1749-8546. (BioMed Central Ltd.)A review. Abstr.: Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) recently was declared a pandemic by world health organization (WHO) Due to sudden outbreaks, currently, no completely effective vaccine or drug is clin. approved. Several therapeutic strategies can be envisaged to prevent further mortality and morbidity. Based on the past contribution of traditional Chinese medicines (TCM) and immune-based therapies as a treatment option in crucial pathogen outbreaks, we aimed to summarize potential therapeutic strategies that could be helpful to stop further spread of SARS-CoV-2 by effecting its structural components or modulation of immune responses. A review. Several TCM with or without modification could be effective against the structural protein, enzymes, and nucleic acid should be tested from available libraries or to identify their immune-stimulatory activities to enhance several antiviral biol. agents for effective elimination of SARS-CoV-2 from the host. TCM is not only effective in the direct inhibition of virus attachment and internalization in a cell but can also prevent their replication and can also help to boost up host immune response. Immune-modulatory effects of TCMs may lead to new medications and can guide us for the scientific validity of drug development. Besides, we also summarized the effective therapies in clin. for controlling inflammation. This review will be not only helpful for the current situation of COVID-19, but can also play a major role in such epidemics in the future.
- 8Choudhary, S.; Malik, Y. S.; Tomar, S. Identification of SARS-CoV-2 cell entry inhibitors by drug repurposing using in silico structure-based virtual screening approach. Front. Immunol. 2020, 11, 1664, DOI: 10.3389/fimmu.2020.01664Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlSis7vM&md5=dc9eff66bcebb7f5a900170511eef385Identification of SARS-CoV-2 cell entry inhibitors by drug repurposing using in silico structure-based virtual screening approachChoudhary, Shweta; Malik, Yashpal S.; Tomar, ShaillyFrontiers in Immunology (2020), 11 (), 1664CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)The rapidly spreading, highly contagious and pathogenic SARS-coronavirus 2 (SARS-CoV-2) assocd. Coronavirus Disease 2019 (COVID-19) has been declared as a pandemic by the World Health Organization (WHO). The novel 2019 SARS-CoV-2 enters the host cell by binding of the viral surface spike glycoprotein (S-protein) to cellular angiotensin converting enzyme 2 (ACE2) receptor. The virus specific mol. interaction with the host cell represents a promising therapeutic target for identifying SARS-CoV-2 antiviral drugs. The repurposing of drugs can provide a rapid and potential cure toward exponentially expanding COVID-19. Thereto, high throughput virtual screening approach was used to investigate FDA approved LOPAC library drugs against both the receptor binding domain of spike protein (S-RBD) and ACE2 host cell receptor. Primary screening identified a few promising mols. for both the targets, which were further analyzed in details by their binding energy, binding modes through mol. docking, dynamics and simulations. Evidently, GR 127935 hydrochloride hydrate, GNF-5, RS504393, TNP, and eptifibatide acetate were found binding to virus binding motifs of ACE2 receptor. Addnl., KT203, BMS195614, KT185, RS504393, and GSK1838705A were identified to bind at the receptor binding site on the viral S-protein. These identified mols. may effectively assist in controlling the rapid spread of SARS-CoV-2 by not only potentially inhibiting the virus at entry step but are also hypothesized to act as anti-inflammatory agents, which could impart relief in lung inflammation. Timely identification and detn. of an effective drug to combat and tranquilize the COVID-19 global crisis is the utmost need of hour. Further, prompt in vivo testing to validate the anti-SARS-CoV-2 inhibition efficiency by these mols. could save lives is justified.
- 9Shyr, Z. A.; Gorshkov, K.; Chen, C. Z.; Zheng, W. Drug discovery strategies for SARS-CoV-2. J. Pharmacol. Exp. Ther. 2020, 375, 127– 138, DOI: 10.1124/jpet.120.000123Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1Kis7jE&md5=c0bfab352eefb70fe2b137a8b7d3899fDrug discovery strategies for SARS-CoV-2Shyr, Zeenat A.; Gorshkov, Kirill; Chen, Catherine Z.; Zheng, WeiJournal of Pharmacology and Experimental Therapeutics (2020), 375 (1), 127-138CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)A review. Coronavirus disease 2019 (COVID-19) is a novel disease caused by the severe acute respiratory syndrome coronavirus (SARS-CoV)-2 virus that was first detected in Dec. of 2019 in Wuhan, China, and has rapidly spread worldwide. The search for a suitable vaccine as well as effective therapeutics for the treatment of COVID-19 is underway. Drug repurposing screens provide a useful and effective soln. for identifying potential therapeutics against SARS-CoV-2. For example, the exptl. drug remdesivir, originally developed for Ebola virus infections, has been approved by the US Food and Drug Administration as an emergency use treatment of COVID-19. However, the efficacy and toxicity of this drug need further improvements. In this review, we discuss recent findings on the pathol. of coronaviruses and the drug targets for the treatment of COVID-19. Both SARS-CoV-2-specific inhibitors and broad-spectrum anticoronavirus drugs against SARS-CoV, Middle East respiratory syndrome coronavirus, and SARS-CoV-2 will be valuable addns. to the anti-SARS-CoV-2 armament. A multitarget treatment approach with synergistic drug combinations contg. different mechanisms of action may be a practical therapeutic strategy for the treatment of severe COVID-19.
- 10Gil, C.; Ginex, T.; Maestro, I.; Nozal, V.; Barrado-Gil, L.; Cuesta-Geijo, M.Á.; Urquiza, J.; Ramírez, D.; Alonso, C.; Campillo, N. E.; Martinez, A. COVID-19: drug targets and potential treatments. J. Med. Chem. 2020, 63, 12359– 12386, DOI: 10.1021/acs.jmedchem.0c00606Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFWmsL3I&md5=6a0dc17c5f601f09a92da933f81bbbeaCOVID-19: Drug Targets and Potential TreatmentsGil, Carmen; Ginex, Tiziana; Maestro, Ines; Nozal, Vanesa; Barrado-Gil, Lucia; Cuesta-Geijo, Miguel Angel; Urquiza, Jesus; Ramirez, David; Alonso, Covadonga; Campillo, Nuria E.; Martinez, AnaJournal of Medicinal Chemistry (2020), 63 (21), 12359-12386CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Currently, the authors are immersed in a pandemic caused by the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which severely threatens public health worldwide. Until now, no drug or vaccine has been approved to treat the severe disease caused by this coronavirus, COVID-19. The authors will focus on the main virus-based and host-based targets that can guide medicinal chem. efforts to discover new drugs for this devastating disease. In principle, all CoVs enzymes and proteins involved in viral replication and the control of host cellular machineries are potentially druggable targets in the search for therapeutic options for SARS-CoV-2. This perspective provides an overview of the main targets from a structural point of view, together with reported therapeutic compds. with activity against SARS-CoV-2 and/or other CoVs. Also, the role of innate immune response to coronavirus infection and the related therapeutic options will be presented.
- 11Catanzaro, M.; Fagiani, F.; Racchi, M.; Corsini, E.; Govoni, S.; Lanni, C. Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transduct Target Ther. 2020, 5, 84, DOI: 10.1038/s41392-020-0191-1Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38rivVOmuw%253D%253D&md5=3fb8e4f85321d6225c5c4922661eab10Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2Catanzaro Michele; Fagiani Francesca; Racchi Marco; Govoni Stefano; Lanni Cristina; Fagiani Francesca; Corsini EmanuelaSignal transduction and targeted therapy (2020), 5 (1), 84 ISSN:.To date, no vaccines or effective drugs have been approved to prevent or treat COVID-19 and the current standard care relies on supportive treatments. Therefore, based on the fast and global spread of the virus, urgent investigations are warranted in order to develop preventive and therapeutic drugs. In this regard, treatments addressing the immunopathology of SARS-CoV-2 infection have become a major focus. Notably, while a rapid and well-coordinated immune response represents the first line of defense against viral infection, excessive inflammatory innate response and impaired adaptive host immune defense may lead to tissue damage both at the site of virus entry and at systemic level. Several studies highlight relevant changes occurring both in innate and adaptive immune system in COVID-19 patients. In particular, the massive cytokine and chemokine release, the so-called "cytokine storm", clearly reflects a widespread uncontrolled dysregulation of the host immune defense. Although the prospective of counteracting cytokine storm is compelling, a major limitation relies on the limited understanding of the immune signaling pathways triggered by SARS-CoV-2 infection. The identification of signaling pathways altered during viral infections may help to unravel the most relevant molecular cascades implicated in biological processes mediating viral infections and to unveil key molecular players that may be targeted. Thus, given the key role of the immune system in COVID-19, a deeper understanding of the mechanism behind the immune dysregulation might give us clues for the clinical management of the severe cases and for preventing the transition from mild to severe stages.
- 12Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X.; Zheng, M.; Chen, L.; Li, H. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm. Sin. B 2020, 10, 766– 788, DOI: 10.1016/j.apsb.2020.02.008Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnslWnug%253D%253D&md5=e644ad7f22b6780443864db76255c8fcAnalysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methodsWu, Canrong; Liu, Yang; Yang, Yueying; Zhang, Peng; Zhong, Wu; Wang, Yali; Wang, Qiqi; Xu, Yang; Li, Mingxue; Li, Xingzhou; Zheng, Mengzhu; Chen, Lixia; Li, HuaActa Pharmaceutica Sinica B (2020), 10 (5), 766-788CODEN: APSBCW; ISSN:2211-3835. (Elsevier B.V.)SARS-CoV-2 has caused tens of thousands of infections and more than one thousand deaths. There are currently no registered therapies for treating coronavirus infections. Because of time consuming process of new drug development, drug repositioning may be the only soln. to the epidemic of sudden infectious diseases. We systematically analyzed all the proteins encoded by SARS-CoV-2 genes, compared them with proteins from other coronaviruses, predicted their structures, and built 19 structures that could be done by homol. modeling. By performing target-based virtual ligand screening, a total of 21 targets (including two human targets) were screened against compd. libraries including ZINC drug database and our own database of natural products. Structure and screening results of important targets such as 3-chymotrypsin-like protease (3CLpro), Spike, RNA-dependent RNA polymerase (RdRp), and papain like protease (PLpro) were discussed in detail. In addn., a database of 78 commonly used anti-viral drugs including those currently on the market and undergoing clin. trials for SARS-CoV-2 was constructed. Possible targets of these compds. and potential drugs acting on a certain target were predicted. This study will provide new lead compds. and targets for further in vitro and in vivo studies of SARS-CoV-2, new insights for those drugs currently ongoing clin. studies, and also possible new strategies for drug repositioning to treat SARS-CoV-2 infections.
- 13VanPatten, S.; He, M.; Altiti, A.; F Cheng, K.; Ghanem, M. H.; Al-Abed, Y. Evidence supporting the use of peptides and peptidomimetics as potential SARS-CoV-2 (COVID-19) therapeutics. Future Med. Chem. 2020, 12, 1647– 1656, DOI: 10.4155/fmc-2020-0180Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvV2nsLzM&md5=de5929a158d71c2f4f8039ebff146307Evidence supporting the use of peptides and peptidomimetics as potential SARS-CoV-2 (COVID-19) therapeuticsVanPatten, Sonya; He, Mingzhu; Altiti, Ahmad; F Cheng, Kai; Ghanem, Mustafa H.; Al-Abed, YousefFuture Medicinal Chemistry (2020), 12 (18), 1647-1656CODEN: FMCUA7; ISSN:1756-8919. (Newlands Press Ltd.)A review. During a disease outbreak/pandemic situation such as COVID-19, researchers are in a prime position to identify and develop peptide-based therapies, which could be more rapidly and cost-effectively advanced into a clin. setting. One drawback of natural peptide drugs, however, is their proteolytic instability; peptidomimetics can help to overcome this caveat. In this review, we summarize peptide and peptide-based therapeutics that target one main entry pathway of SARS-CoV-2, which involves the host ACE2 receptor and viral spike (S) protein interaction. Furthermore, we discuss the advantages of peptidomimetics and other potential targets that have been studied using peptide-based therapeutics for COVID-19.
- 14Smith, M. C.; Gestwicki, J. E. Features of protein-protein interactions that translate into potent inhibitors: topology, surface area and affinity. Expert Rev. Mol. Med. 2012, 14, 14– 16, DOI: 10.1017/erm.2012.10Google ScholarThere is no corresponding record for this reference.
- 15Josephson, K.; Ricardo, A.; Szostak, J. W. mRNA display: from basic principles to macrocycle drug discovery. Drug Discovery Today 2014, 19, 388– 99, DOI: 10.1016/j.drudis.2013.10.011Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsleltrnI&md5=6676fef2ca8f0a81cc0e993ee4ec7424mRNA display: from basic principles to macrocycle drug discoveryJosephson, Kristopher; Ricardo, Alonso; Szostak, Jack W.Drug Discovery Today (2014), 19 (4), 388-399CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. We describe a new discovery technol. that uses mRNA-display to rapidly synthesize and screen macrocyclic peptide libraries to explore a valuable region of chem. space typified by natural products. This technol. allows high-affinity peptidic macrocycles contg. modified backbones and unnatural side chains to be readily selected based on target binding. Success stories covering the first examples of these libraries suggest that they could be used for the discovery of intracellular protein-protein interaction inhibitors, highly selective enzyme inhibitors or synthetic replacements for monoclonal antibodies. The review concludes with a look to the future regarding how this technol. might be improved with respect to library design for cell permeability and bioavailability.
- 16WHO. Draft landscape of COVID-19 candidate vaccines. https://www.who.int/news-room/q-a-detail/coronavirus-disease-(covid-19)-vaccines (accessed January 2021).Google ScholarThere is no corresponding record for this reference.
- 17Voysey, M.; Clemens; Madhi, S.; Weckx, L. Y.; Folegatti, P. M. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021, 397, 99– 111, DOI: 10.1016/S0140-6736(20)32661-1Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFCgt77I&md5=826fc2243914f0e5bcdf0931520c2b42Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomized controlled trials in Brazil, South Africa, and the UKVoysey, Merryn; Clemens, Sue Ann Costa; Madhi, Shabir A.; Weckx, Lily Y.; Folegatti, Pedro M.; Aley, Parvinder K.; Angus, Brian; Baillie, Vicky L.; Barnabas, Shaun L.; Bhorat, Qasim E.; Bibi, Sagida; Briner, Carmen; Cicconi, Paola; Collins, Andrea M.; Colin-Jones, Rachel; Cutland, Clare L.; Darton, Thomas C.; Dheda, Keertan; Duncan, Christopher J. A.; Emary, Katherine R. W.; Ewer, Katie J.; Fairlie, Lee; Faust, Saul N.; Feng, Shuo; Ferreira, Daniela M.; Finn, Adam; Goodman, Anna L.; Green, Catherine M.; Green, Christopher A.; Heath, Paul T.; Hill, Catherine; Hill, Helen; Hirsch, Ian; Hodgson, Susanne H. C.; Izu, Alane; Jackson, Susan; Jenkin, Daniel; Joe, Carina C. D.; Kerridge, Simon; Koen, Anthonet; Kwatra, Gaurav; Lazarus, Rajeka; Lawrie, Alison M.; Lelliott, Alice; Libri, Vincenzo; Lillie, Patrick J.; Mallory, Raburn; Mendes, Ana V. A.; Milan, Eveline P.; Minassian, Angela M.; McGregor, Alastair; Morrison, Hazel; Mujadidi, Yama F.; Nana, Anusha; O'Reilly, Peter J.; Padayachee, Sherman D.; Pittella, Ana; Plested, Emma; Pollock, Katrina M.; Ramasamy, Maheshi N.; Rhead, Sarah; Schwarzbold, Alexandre V.; Singh, Nisha; Smith, Andrew; Song, Rinn; Snape, Matthew D.; Sprinz, Eduardo; Sutherland, Rebecca K.; Tarrant, Richard; Thomson, Emma C.; Torok, M. Estee; Toshner, Mark; Turner, David P. J.; Vekemans, Johan; Villafana, Tonya L.; Watson, Marion E. E.; Williams, Christopher J.; Douglas, Alexander D.; Hill, Adrian V. S.; Lambe, Teresa; Gilbert, Sarah C.; Pollard, Andrew J.Lancet (2021), 397 (10269), 99-111CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A safe and efficacious vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), if deployed with high coverage, could contribute to the control of the COVID-19 pandemic. We evaluated the safety and efficacy of the ChAdOx1 nCoV-19 vaccine in a pooled interim anal. of four trials. This anal. includes data from four ongoing blinded, randomized, controlled trials done across the UK, Brazil, and South Africa. Participants aged 18 years and older were randomly assigned (1:1) to ChAdOx1 nCoV-19 vaccine or control (meningococcal group A, C, W, and Y conjugate vaccine or saline). Participants in the ChAdOx1 nCoV-19 group received two doses contg. 5 x 1010 viral particles (std. dose; SD/SD cohort); a subset in the UK trial received a half dose as their first dose (low dose) and a std. dose as their second dose (LD/SD cohort). The primary efficacy anal. included symptomatic COVID-19 in seroneg. participants with a nucleic acid amplification test-pos. swab more than 14 days after a second dose of vaccine. Participants were analyzed according to treatment received, with data cutoff on Nov 4, 2020. Vaccine efficacy was calcd. as 1 - relative risk derived from a robust Poisson regression model adjusted for age. Studies are registered at ISRCTN89951424 and ClinicalTrials.gov, NCT04324606, NCT04400838, and NCT04444674. Between Apr. 23 and Nov 4, 2020, 23,848 participants were enrolled and 11,636 participants (7548 in the UK, 4088 in Brazil) were included in the interim primary efficacy anal. In participants who received two std. doses, vaccine efficacy was 62.1% (95% CI 41.0-75.7; 27 [0.6%] of 4440 in the ChAdOx1 nCoV-19 group vs. 71 [1.6%] of 4455 in the control group) and in participants who received a low dose followed by a std. dose, efficacy was 90.0% (67.4-97.0; three [0.2%] of 1367 vs 30 [2.2%] of 1374; pinteraction=0.010). Overall vaccine efficacy across both groups was 70.4% (95.8% CI 54.8-80.6; 30 [0.5%] of 5807 vs 101 [1.7%] of 5829). From 21 days after the first dose, there were ten cases hospitalized for COVID-19, all in the control arm; two were classified as severe COVID-19, including one death. There were 74,341 person-months of safety follow-up (median 3.4 mo, IQR 1.3-4.8): 175 severe adverse events occurred in 168 participants, 84 events in the ChAdOx1 nCoV-19 group and 91 in the control group. Three events were classified as possibly related to a vaccine: one in the ChAdOx1 nCoV-19 group, one in the control group, and one in a participant who remains masked to group allocation. ChAdOx1 nCoV-19 has an acceptable safety profile and has been found to be efficacious against symptomatic COVID-19 in this interim anal. of ongoing clin. trials.
- 18Polack, F. P.; Thomas, S. J.; Kitchin, N. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N. Engl. J. Med. 2020, 383, 2603– 2615, DOI: 10.1056/NEJMoa2034577Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFSjuw%253D%253D&md5=08cb7204334ed37c7949dfcdfe5af75cSafety and efficacy of the BNT162b2 mRNA Covid-19 vaccinePolack, Fernando P.; Thomas, Stephen J.; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Perez, John L.; Marc, Gonzalo Perez; Moreira, Edson D.; Zerbini, Cristiano; Bailey, Ruth; Swanson, Kena A.; Roychoudhury, Satrajit; Koury, Kenneth; Li, Ping; Kalina, Warren V.; Cooper, David; Frenck, Robert W., Jr.; Hammitt, Laura L.; Tureci, Ozlem; Nell, Haylene; Schaefer, Axel; Unal, Serhat; Tresnan, Dina B.; Mather, Susan; Dormitzer, Philip R.; Sahin, Ugur; Jansen, Kathrin U.; Gruber, William C.New England Journal of Medicine (2020), 383 (27), 2603-2615CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)A review. Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a worldwide pandemic. Safe and effective vaccines are needed urgently. methods In an ongoing multinational, placebo-controlled, observer-blinded, pivotal efficacy trial, we randomly assigned persons 16 years of age or older in a 1:1 ratio to receive two doses, 21 days apart, of either placebo or the BNT162b2 vaccine candidate (30μg per dose). BNT162b2 is a lipid nanoparticle-formulated, nucleoside-modified RNA vaccine that encodes a prefusion stabilized, membrane-anchored SARS-CoV-2 full-length spike protein. The primary end points were efficacy of the vaccine against lab.-confirmed Covid-19 and safety. results A total of 43,548 participants underwent randomization, of whom 43,448 received injections: 21,720 with BNT162b2 and 21,728 with placebo. There were 8 cases of Covid-19 with onset at least 7 days after the second dose among participants assigned to receive BNT162b2 and 162 cases among those assigned to placebo; BNT162b2 was 95% effective in preventing Covid-19 (95% credible interval, 90.3 to 97.6). Similar vaccine efficacy (generally 90 to 100%) was obsd. across subgroups defined by age, sex, race, ethnicity, baseline body-mass index, and the presence of coexisting conditions. Among 10 cases of severe Covid-19 with onset after the first dose, 9 occurred in placebo recipients and 1 in a BNT162b2 recipient. The safety profile of BNT162b2 was characterized by short-term, mild-to-moderate pain at the injection site, fatigue, and headache. The incidence of serious adverse events was low and was similar in the vaccine and placebo groups. conclusions A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older. Safety over a median of 2 mo was similar to that of other viral vaccines.
- 19Baden, L. R.; El Sahly, H. M.; Essink, B. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 2021, 384, 403– 416, DOI: 10.1056/NEJMoa2035389Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFSmsrc%253D&md5=9b0c79f3e3fd2ea452a3fbf57281720fEfficacy and safety of the mRNA-1273 SARS-CoV-2 vaccineBaden, L. R.; El Sahly, H. M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S. A.; Rouphael, N.; Creech, C. B.; McGettigan, J.; Khetan, S.; Segall, N.; Solis, J.; Brosz, A.; Fierro, C.; Schwartz, H.; Neuzil, K.; Corey, L.; Gilbert, P.; Janes, H.; Follmann, D.; Marovich, M.; Mascola, J.; Polakowski, L.; Ledgerwood, J.; Graham, B. S.; Bennett, H.; Pajon, R.; Knightly, C.; Leav, B.; Deng, W.; Zhou, H.; Han, S.; Ivarsson, M.; Miller, J.; Zaks, T.New England Journal of Medicine (2021), 384 (5), 403-416CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Vaccines are needed to prevent coronavirus disease 2019 (Covid-19) and to protect persons who are at high risk for complications. The mRNA-1273 vaccine is a lipid nanoparticle-encapsulated mRNA-based vaccine that encodes the prefusion stabilized full-length spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes Covid-19. This phase 3 randomized, observer-blinded, placebo-controlled trial was conducted at 99 centers across the United States. Persons at high risk for SARS-CoV-2 infection or its complications were randomly assigned in a 1:1 ratio to receive two i.m. injections of mRNA-1273 (100μg) or placebo 28 days apart. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second injection in participants who had not previously been infected with SARS-CoV-2. The trial enrolled 30,420 volunteers who were randomly assigned in a 1:1 ratio to receive either vaccine or placebo (15,210 participants in each group). More than 96% of participants received both injections, and 2.2% had evidence (serol., virol., or both) of SARS-CoV-2 infection at baseline. Symptomatic Covid-19 illness was confirmed in 185 participants in the placebo group (56.5 per 1000 person-years; 95% confidence interval [CI], 48.7 to 65.3) and in 11 participants in the mRNA-1273 group (3.3 per 1000 person-years; 95% CI, 1.7 to 6.0); vaccine efficacy was 94.1% (95% CI, 89.3 to 96.8%; P<0.001). Efficacy was similar across key secondary analyses, including assessment 14 days after the first dose, analyses that included participants who had evidence of SARS-CoV-2 infection at baseline, and analyses in participants 65 years of age or older. Severe Covid-19 occurred in 30 participants, with one fatality; all 30 were in the placebo group. Moderate, transient reactogenicity after vaccination occurred more frequently in the mRNA-1273 group. Serious adverse events were rare, and the incidence was similar in the two groups. The mRNA-1273 vaccine showed 94.1% efficacy at preventing Covid-19 illness, including severe disease. Aside from transient local and systemic reactions, no safety concerns were identified.
- 20Kovyrshina, A. V.; Dolzhikova, I. V.; Grousova, D. M. A heterologous virus-vectored vaccine for prevention of middle east respiratory syndrome induces long protective immune response against MERS-CoV. Immunologiya 2020, 41, 135– 43, DOI: 10.33029/0206-4952-2020-41-2-135-143Google ScholarThere is no corresponding record for this reference.
- 21Logunov, D. Y.; Dolzhikova, I. V.; Zubkova, O. V. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet 2020, 396, 887– 97, DOI: 10.1016/S0140-6736(20)31866-3Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsl2ktrfF&md5=68963e757a4869be9a28726203eacc8bSafety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from RussiaLogunov, Denis Y.; Dolzhikova, Inna V.; Zubkova, Olga V.; Tukhvatullin, Amir I.; Shcheblyakov, Dmitry V.; Dzharullaeva, Alina S.; Grousova, Daria M.; Erokhova, Alina S.; Kovyrshina, Anna V.; Botikov, Andrei G.; Izhaeva, Fatima M.; Popova, Olga; Ozharovskaya, Tatiana A.; Esmagambetov, Ilias B.; Favorskaya, Irina A.; Zrelkin, Denis I.; Voronina, Daria V.; Shcherbinin, Dmitry N.; Semikhin, Alexander S.; Simakova, Yana V.; Tokarskaya, Elizaveta A.; Lubenets, Nadezhda L.; Egorova, Daria A.; Shmarov, Maksim M.; Nikitenko, Natalia A.; Morozova, Lola F.; Smolyarchuk, Elena A.; Kryukov, Evgeny V.; Babira, Vladimir F.; Borisevich, Sergei V.; Naroditsky, Boris S.; Gintsburg, Alexander L.Lancet (2020), 396 (10255), 887-897CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)We developed a heterologous COVID-19 vaccine consisting of 2 components, a recombinant adenovirus type 26 (rAd26) vector and a recombinant adenovirus type 5 (rAd5) vector, both carrying the gene for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein (rAd26-S and rAd5-S). We aimed to assess the safety and immunogenicity of 2 formulations (frozen and lyophilized) of this vaccine. We did 2 open, non-randomized phase 1/2 studies at 2 hospitals in Russia. We enrolled healthy adult volunteers (men and women) aged 18-60 yr to both studies. In phase 1 of each study, we administered i.m. on day 0 either 1 dose of rAd26-S or 1 dose of rAd5-S and assessed the safety of the 2 components for 28 days. In phase 2 of the study, which began no earlier than 5 days after phase 1 vaccination, we administered i.m. a prime-boost vaccination, with rAd26-S given on day 0 and rAd5-S on day 21. Primary outcome measures were antigen-specific humoral immunity (SARS-CoV-2-specific antibodies measured by ELISA on days 0, 14, 21, 28, and 42) and safety (no. of participants with adverse events monitored throughout the study). Secondary outcome measures were antigen-specific cellular immunity (T-cell responses and interferon-γ concn.) and change in neutralizing antibodies (detected with a SARS-CoV-2 neutralization assay). These trials are registered with ClinicalTrials.gov, NCT04436471 and NCT04437875. Between June 18 and Aug 3, 2020, we enrolled 76 participants to the 2 studies (38 in each study). In each study, 9 volunteers received rAd26-S in phase 1, 9 received rAd5-S in phase 1, and 20 received rAd26-S and rAd5-S in phase 2. Both vaccine formulations were safe and well tolerated. The most common adverse events were pain at injection site (44 [58%]), hyperthermia (38 [50%]), headache (32 [42%]), asthenia (21 [28%]), and muscle and joint pain (18 [24%]). Most adverse events were mild and no serious adverse events were detected. All participants produced antibodies to SARS-CoV-2 glycoprotein. At day 42, receptor binding domain-specific IgG titers were 14 703 with the frozen formulation and 11,143 with the lyophilized formulation, and neutralizing antibodies were 49·25 with the frozen formulation and 45·95 with the lyophilized formulation, with a seroconversion rate of 100%. Cell-mediated responses were detected in all participants at day 28, with median cell proliferation of 2.5% CD4+ and 1.3% CD8+ with the frozen formulation, and a median cell proliferation of 1.3% CD4+ and 1.1% CD8+ with the lyophilized formulation. The heterologous rAd26 and rAd5 vector-based COVID-19 vaccine has a good safety profile and induced strong humoral and cellular immune responses in participants. Further investigation is needed of the effectiveness of this vaccine for prevention of COVID-19. Ministry of Health of the Russian Federation.
- 22Logunov, D. Y.; Dolzhikova, I. V.; Shcheblyakov, D. V.; Tukhvatulin, A. Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomised controlled phase 3 trial in Russia. Lancet 2021, 397, 671, DOI: 10.1016/S0140-6736(21)00234-8Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjslajtrs%253D&md5=94062efdc9d84abcfe49ebae871934a4Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomized controlled phase 3 trial in RussiaLogunov, Denis Y.; Dolzhikova, Inna V.; Shcheblyakov, Dmitry V.; Tukhvatulin, Amir I.; Zubkova, Olga V.; Dzharullaeva, Alina S.; Kovyrshina, Anna V.; Lubenets, Nadezhda L.; Grousova, Daria M.; Erokhova, Alina S.; Botikov, Andrei G.; Izhaeva, Fatima M.; Popova, Olga; Ozharovskaya, Tatiana A.; Esmagambetov, Ilias B.; Favorskaya, Irina A.; Zrelkin, Denis I.; Voronina, Daria V.; Shcherbinin, Dmitry N.; Semikhin, Alexander S.; Simakova, Yana V.; Tokarskaya, Elizaveta A.; Egorova, Daria A.; Shmarov, Maksim M.; Nikitenko, Natalia A.; Gushchin, Vladimir A.; Smolyarchuk, Elena A.; Zyryanov, Sergey K.; Borisevich, Sergei V.; Naroditsky, Boris S.; Gintsburg, Alexander L.Lancet (2021), 397 (10275), 671-681CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A heterologous recombinant adenovirus (rAd)-based vaccine, Gam-COVID-Vac (Sputnik V), showed a good safety profile and induced strong humoral and cellular immune responses in participants in phase 1/2 clin. trials. Here, we report preliminary results on the efficacy and safety of Gam-COVID-Vac from the interim anal. of this phase 3 trial. We did a randomized, double-blind, placebo-controlled, phase 3 trial at 25 hospitals and polyclinics in Moscow, Russia. We included participants aged at least 18 years, with neg. SARS-CoV-2 PCR and IgG and IgM tests, no infectious diseases in the 14 days before enrollment, and no other vaccinations in the 30 days before enrollment. Participants were randomly assigned (3:1) to receive vaccine or placebo, with stratification by age group. Investigators, participants, and all study staff were masked to group assignment. The vaccine was administered (0·5 mL/dose) i.m. in a prime-boost regimen: a 21-day interval between the first dose (rAd26) and the second dose (rAd5), both vectors carrying the gene for the full-length SARS-CoV-2 glycoprotein S. The primary outcome was the proportion of participants with PCR-confirmed COVID-19 from day 21 after receiving the first dose. All analyses excluded participants with protocol violations: the primary outcome was assessed in participants who had received two doses of vaccine or placebo, serious adverse events were assessed in all participants who had received at least one dose at the time of database lock, and rare adverse events were assessed in all participants who had received two doses and for whom all available data were verified in the case report form at the time of database lock. The trial is registered at ClinicalTrials.gov (NCT04530396). Between Sept 7 and Nov 24, 2020, 21 977 adults were randomly assigned to the vaccine group (n=16 501) or the placebo group (n=5476). 19 866 received two doses of vaccine or placebo and were included in the primary outcome anal. From 21 days after the first dose of vaccine (the day of dose 2), 16 (0·1%) of 14 964 participants in the vaccine group and 62 (1·3%) of 4902 in the placebo group were confirmed to have COVID-19; vaccine efficacy was 91·6% (95% CI 85·6-95·2). Most reported adverse events were grade 1 (7485 [94·0%] of 7966 total events). 45 (0·3%) of 16 427 participants in the vaccine group and 23 (0·4%) of 5435 participants in the placebo group had serious adverse events; none were considered assocd. with vaccination, with confirmation from the independent data monitoring committee. Four deaths were reported during the study (three [<0·1%] of 16 427 participants in the vaccine group and one [<0·1%] of 5435 participants in the placebo group), none of which were considered related to the vaccine. This interim anal. of the phase 3 trial of Gam-COVID-Vac showed 91·6% efficacy against COVID-19 and was well tolerated in a large cohort. Moscow City Health Department, Russian Direct Investment Fund, Sberbank, and RUSAL.
- 23WHO. Transmission of SARS-CoV-2: implications for infection prevention precautions. https://www.who.int/news-room/commentaries/detail/transmission-of-sars-cov-2-implications-for-infection-prevention-precautions (accessed January 2020).Google ScholarThere is no corresponding record for this reference.
- 24Centers for disease control and prevention. Interim clinical guidance for management of patients with confirmed coronavirus disease (COVID-19). https://www.cdc.gov/coronavirus/2019-ncov/hcp/ (accessed May 2020).Google ScholarThere is no corresponding record for this reference.
- 25WHO. Target product profiles for COVID-19 vaccines , 2020. https://www.who.int/publications/m/item/who-target-product-profiles-for-covid-19-vaccines (accessed January 2020).Google ScholarThere is no corresponding record for this reference.
- 26Amanat, F. K. SARS-CoV-2 vaccines: status report. Immunity 2020, 52, 583, DOI: 10.1016/j.immuni.2020.03.007Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXmsVygs7k%253D&md5=63071e7666cff2dd50c48fc51cdf3c99SARS-CoV-2 Vaccines: Status ReportAmanat, Fatima; Krammer, FlorianImmunity (2020), 52 (4), 583-589CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)A review. SARS-CoV-2, the causal agent of COVID-19, first emerged in late 2019 in China. It has since infected more than 870,000 individuals and caused more than 43,000 deaths globally. Here, we discuss therapeutic and prophylactic interventions for SARS-CoV-2 with a focus on vaccine development and its challenges. Vaccines are being rapidly developed but will likely come too late to affect the first wave of a potential pandemic. Nevertheless, crit. lessons can be learned for the development of vaccines against rapidly emerging viruses. Importantly, SARS-CoV-2 vaccines will be essential to reducing morbidity and mortality if the virus establishes itself in the population.
- 27Chen, W. H.; Strych, U.; Hotez, P. J.; Bottazzi, M. E. The SARS-CoV-2 vaccine pipeline: an overview. Curr. Trop Med. Rep. 2020, 7, 61, DOI: 10.1007/s40475-020-00201-6Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB383otFWmtg%253D%253D&md5=10552f461e781e5eb4567cd3c14060ecThe SARS-CoV-2 Vaccine Pipeline: an OverviewChen Wen-Hsiang; Strych Ulrich; Hotez Peter J; Bottazzi Maria ElenaCurrent tropical medicine reports (2020), 7 (2), 61-64 ISSN:2196-3045.Purpose of Review: The goal of this review is to provide a timely overview on efforts to develop a vaccine for the 2019 novel coronavirus SARS-CoV-2, the causative agent of coronavirus disease (COVID-19). Recent Findings: Previous research efforts to develop a severe acute respiratory syndrome coronavirus (SARS-CoV) vaccine in the years following the 2003 pandemic have opened the door for investigators to design vaccine concepts and approaches for the COVID-19 epidemic in China. Both SARS-CoV and SARS-CoV-2 exhibit a high degree of genetic similarity and bind to the same host cell ACE2 receptor. Based on previous experience with SARS-CoV vaccines, it is expected that all COVID-19 vaccines will require careful safety evaluations for immunopotentiation that could lead to increased infectivity or eosinophilic infiltration. Besides this, a COVID-19 vaccine target product profile must address vaccinating at-risk human populations including frontline healthcare workers, individuals over the age of 60, and those with underlying and debilitating chronic conditions. Among the vaccine technologies under evaluation are whole virus vaccines, recombinant protein subunit vaccines, and nucleic acid vaccines. Summary: Each current vaccine strategy has distinct advantages and disadvantages. Therefore, it is paramount that multiple strategies be advanced quickly and then evaluated for safety and efficacy. Ultimately, the safety studies to minimize undesired immunopotentiation will become the most significant bottleneck in terms of time.
- 28Leader, B.; Baca, Q. J.; Golan, D. E. Protein therapeutics: a summary and pharmacological classification. Nat. Rev. Drug Discovery 2008, 7, 21– 39, DOI: 10.1038/nrd2399Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1ygtQ%253D%253D&md5=761a9860e38c98295385641dda94b31fProtein therapeutics: a summary and pharmacological classificationLeader, Benjamin; Baca, Quentin J.; Golan, David E.Nature Reviews Drug Discovery (2008), 7 (1), 21-39CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Once a rarely used subset of medical treatments, protein therapeutics have increased dramatically in no. and frequency of use since the introduction of the first recombinant protein therapeutic - human insulin - 25 years ago. Protein therapeutics already have a significant role in almost every field of medicine, but this role is still only in its infancy. This article overviews some of the key characteristics of protein therapeutics, summarizes the more than 130 protein therapeutics used currently and suggests a new classification of these proteins according to their pharmacol. action.
- 29Suntharalingam, G.; Perry, M. R.; Ward, S.; Brett, S. J.; Panoskaltsis, N. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 2006, 355, 1018– 1028, DOI: 10.1056/NEJMoa063842Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpsFKmtbc%253D&md5=e2d2ace0459e5f0d7472de79e68ae37eCytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412Suntharalingam, Ganesh; Perry, Meghan R.; Ward, Stephen; Brett, Stephen J.; Castello-Cortes, Andrew; Brunner, Michael D.; Panoskaltsis, NickiNew England Journal of Medicine (2006), 355 (10), 1018-1028CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)Six healthy young male volunteers at a contract research organization were enrolled in the first phase 1 clin. trial of TGN1412, a novel superagonist anti-CD28 monoclonal antibody that directly stimulates T cells. Within 90 min after receiving a single i.v. dose of the drug, all six volunteers had a systemic inflammatory response characterized by a rapid induction of proinflammatory cytokines and accompanied by headache, myalgias, nausea, diarrhea, erythema, vasodilatation, and hypotension. Within 12 to 16 h after infusion, they became critically ill, with pulmonary infiltrates and lung injury, renal failure, and disseminated intravascular coagulation. Severe and unexpected depletion of lymphocytes and monocytes occurred within 24 h after infusion. All six patients were transferred to the care of the authors at an intensive care unit at a public hospital, where they received intensive cardiopulmonary support (including dialysis), high-dose methylprednisolone, and an anti-interleukin-2 receptor antagonist antibody. Prolonged cardiovascular shock and acute respiratory distress syndrome developed in two patients, who required intensive organ support for 8 and 16 days. Despite evidence of the multiple cytokine-release syndrome, all six patients survived. Documentation of the clin. course occurring over the 30 days after infusion offers insight into the systemic inflammatory response syndrome in the absence of contaminating pathogens, endotoxin, or underlying disease.
- 30Wadman, M. London’s disastrous drug trial has serious side effects for research. Nature 2006, 440, 388– 389, DOI: 10.1038/440388aGoogle Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xis1Omt7c%253D&md5=2516c376286b52865e6371181688bf14London's disastrous drug trial has serious side effects for researchWadman, MeredithNature (London, United Kingdom) (2006), 440 (7083), 388-389CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)There is no expanded citation for this reference.
- 31Huck, B. R.; Kötzner, L.; Urbahns, K. Small molecules drive big improvements in immuno-oncology therapies. Angew. Chem., Int. Ed. 2018, 57, 4412– 4428, DOI: 10.1002/anie.201707816Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjtlWqsbo%253D&md5=bef3a381977686b5bdb432a76364b066Small Molecules Drive Big Improvements in Immuno-Oncology TherapiesHuck, Bayard R.; Koetzner, Lisa; Urbahns, KlausAngewandte Chemie, International Edition (2018), 57 (16), 4412-4428CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Immunooncol. therapies have the potential to revolutionize the armamentarium of available cancer treatments. To further improve clin. response rates, researchers are looking for novel combination regimens, with checkpoint blockade being used as a backbone of the treatment. This Review highlights the significance of small mols. in this approach, which holds promise to provide increased benefit to cancer patients.
- 32Gadek, T. R.; Burdick, D. J.; McDowell, R. S.; Stanley, M. S. Generation of an LFA-1 antagonist by the transfer of the ICAM-1 immunoregulatory epitope to a small molecule. Science 2002, 295, 1086– 1089, DOI: 10.1126/science.295.5557.1086Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xht1Gqt7s%253D&md5=bbcb9a2fe2c245e038dd7ddb28462234Generation of an LFA-1 antagonist by the transfer of the ICAM-1 immunoregulatory epitope to a small moleculeGadek, T. R.; Burdick, D. J.; McDowell, R. S.; Stanley, M. S.; Marsters, J. C., Jr.; Paris, K. J.; Oare, D. A.; Reynolds, M. E.; Ladner, C.; Zioncheck, K. A.; Lee, W. P.; Gribling, P.; Dennis, M. S.; Skelton, N. J.; Tumas, D. B.; Clark, K. R.; Keating, S. M.; Beresini, M. H.; Tilley, J. W.; Presta, L. G.; Bodary, S. C.Science (Washington, DC, United States) (2002), 295 (5557), 1086-1089CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The protein-protein interaction between leukocyte functional antigen-1 (LFA-1) and intercellular adhesion mol.-1 (ICAM-1) is crit. to lymphocyte and immune system function. Here, we report on the transfer of the contiguous, nonlinear epitope of ICAM-1, responsible for its assocn. with LFA-1, to a small-mol. framework. These LFA-1 antagonists bound LFA-1, blocked binding of ICAM-1, and inhibited a mixed lymphocyte reaction (MLR) with potency significantly greater than that of cyclosporine A. Furthermore, in comparison to an antibody to LFA-1, they exhibited significant anti-inflammatory effects in vivo. These results demonstrate the utility of small-mol. mimics of nonlinear protein epitopes and the protein epitopes themselves as leads in the identification of novel pharmaceutical agents.
- 33Scott, D. E.; Bayly, A. R.; Abell, C.; Skidmore, J. Small molecules, big targets: drug discovery faces the protein-protein interaction challenge. Nat. Rev. Drug Discovery 2016, 15, 533– 550, DOI: 10.1038/nrd.2016.29Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XlsFOgur4%253D&md5=9f0953567ae9338b8b36aa78b8160655Small molecules, big targets: drug discovery faces the protein-protein interaction challengeScott, Duncan E.; Bayly, Andrew R.; Abell, Chris; Skidmore, JohnNature Reviews Drug Discovery (2016), 15 (8), 533-550CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)Protein-protein interactions (PPIs) are of pivotal importance in the regulation of biol. systems and are consequently implicated in the development of disease states. Recent work has begun to show that, with the right tools, certain classes of PPI can yield to the efforts of medicinal chemists to develop inhibitors, and the first PPI inhibitors have reached clin. development. In this Review, we describe the research leading to these breakthroughs and highlight the existence of groups of structurally related PPIs within the PPI target class. For each of these groups, we use examples of successful discovery efforts to illustrate the research strategies that have proved most useful.
- 34Zhang, G.; Pomplun, S.; Loftis, A. R.; Loas, A.; Pentelute, B. L. Investigation of ACE2 N-terminal fragments binding to SARS-CoV-2 spike RBD. bioRxiv , June 17, 2020, ver. 1. DOI: 10.1101/2020.03.19.999318 .Google ScholarThere is no corresponding record for this reference.
- 35Han, Y.; Král, P. Computational design of ACE2-based peptide inhibitors of SARS-CoV-2. ACS Nano 2020, 14, 5143– 5147, DOI: 10.1021/acsnano.0c02857Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXntVKns7s%253D&md5=54088dfed751aff6a9e8dcec95bd25edComputational design of ACE2-based peptide inhibitors of SARS-CoV-2Han, Yanxiao; Kral, PetrACS Nano (2020), 14 (4), 5143-5147CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Peptide inhibitors against the SARS-CoV-2 coronavirus, currently causing a worldwide pandemic, are designed and simulated. The inhibitors are mostly formed by two sequential self-supporting α-helixes (bundle) extd. from the protease domain (PD) of angiotensin-converting enzyme 2 (ACE2), which bind to the SARS-CoV-2 receptor binding domains. Mol. dynamics simulations revealed that the α-helical peptides maintain their secondary structure and provide a highly specific and stable binding (blocking) to SARS-CoV-2. To provide a multivalent binding to the SARS-CoV-2 receptors, many such peptides could be attached to the surfaces of nanoparticle carriers. The proposed peptide inhibitors could provide simple and efficient therapeutics against the COVID-19 disease.
- 36Karoyan, P.; Vieillard, V.; Gómez-Morales, L. Human ACE2 peptide-mimics block SARS-CoV-2 pulmonary cells infection. Commun. Biol. 2021, 4, 197, DOI: 10.1038/s42003-021-01736-8Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktlajsro%253D&md5=4750200c747ebdaab86ff52b6cf0e66aHuman ACE2 peptide-mimics block SARS-CoV-2 pulmonary cells infectionKaroyan, Philippe; Vieillard, Vincent; Gomez-Morales, Luis; Odile, Estelle; Guihot, Amelie; Luyt, Charles-Edouard; Denis, Alexis; Grondin, Pascal; Lequin, OlivierCommunications Biology (2021), 4 (1), 197CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)In light of the recent accumulated knowledge on SARS-CoV-2 and its mode of human cells invasion, the binding of viral spike glycoprotein to human Angiotensin Converting Enzyme 2 (hACE2) receptor plays a central role in cell entry. We designed a series of peptides mimicking the N-terminal helix of hACE2 protein which contains most of the contacting residues at the binding site, exhibiting a high helical folding propensity in aq. soln. Our best peptide-mimics are able to block SARS-CoV-2 human pulmonary cell infection with an inhibitory concn. (IC50) in the nanomolar range upon binding to the virus spike protein with high affinity. These first-in-class blocking peptide mimics represent powerful tools that might be used in prophylactic and therapeutic approaches to fight the coronavirus disease 2019 (COVID-19).
- 37Curreli, F.; Victor, S. M. B.; Ahmed, S.; Drelich, A. Stapled peptides based on human angiotensin-converting enzyme 2 (ACE2) potently inhibit SARS-CoV-2 infection in vitro. mBio 2020, 11, e02451-20 DOI: 10.1128/mBio.02451-20Google ScholarThere is no corresponding record for this reference.
- 38Tai, W. Identification of SARS-CoV RBD-targeting monoclonal antibodies with cross-reactive or neutralizing activity against SARS-CoV-2. Antiviral Res. 2020, 179, 104820, DOI: 10.1016/j.antiviral.2020.104820Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpvFansb8%253D&md5=3418b1f745d3d23944bb6acef604a99dIdentification of SARS-CoV RBD-targeting monoclonal antibodies with cross-reactive or neutralizing activity against SARS-CoV-2Tai, Wanbo; Zhang, Xiujuan; He, Yuxian; Jiang, Shibo; Du, LanyingAntiviral Research (2020), 179 (), 104820CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)SARS-CoV-2-caused COVID-19 cases are growing globally, calling for developing effective therapeutics to control the current pandemic. SARS-CoV-2 and SARS-CoV recognize angiotensin-converting enzyme 2 (ACE2) receptor via the receptor-binding domain (RBD). Here, we identified six SARS-CoV RBD-specific neutralizing monoclonal antibodies (nAbs) that cross-reacted with SARS-CoV-2 RBD, two of which, 18F3 and 7B11, neutralized SARS-CoV-2 infection. 18F3 recognized conserved epitopes on SARS-CoV and SARS-CoV-2 RBDs, whereas 7B11 recognized epitopes on SARS-CoV RBD not fully conserved in SARS-CoV-2 RBD. The 18F3-recognizing epitopes on RBD did not overlap with the ACE2-binding sites, whereas those recognized by 7B11 were close to the ACE2-binding sites, explaining why 7B11 could, but 18F3 could not, block SARS-CoV or SARS-CoV-2 RBD binding to ACE2 receptor. Our study provides an alternative approach to prevent SARS-CoV-2 infection using anti-SARS-CoV nAbs.
- 39De Vries, R. D.; Schmitz, K. S.; Bovier, F. T. Intranasal fusion inhibitory lipopeptide prevents direct contact SARS-CoV-2 transmission in ferrets. bioRxiv , November 5, 2020, ver. 1. DOI: 10.1101/2020.11.04.361154 .Google ScholarThere is no corresponding record for this reference.
- 40Tai, W.; He, L.; Zhang, X.; Pu, J.; Voronin, D.; Jiang, S.; Zhou, Y.; Du, L. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol. Immunol. 2020, 17, 613– 620, DOI: 10.1038/s41423-020-0400-4Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlt1Chsrw%253D&md5=87bc49d070c84e78b01230518aaa465aCharacterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccineTai, Wanbo; He, Lei; Zhang, Xiujuan; Pu, Jing; Voronin, Denis; Jiang, Shibo; Zhou, Yusen; Du, LanyingCellular & Molecular Immunology (2020), 17 (6), 613-620CODEN: CMIEAO; ISSN:1672-7681. (Nature Research)The outbreak of Coronavirus Disease 2019 (COVID-19) has posed a serious threat to global public health, calling for the development of safe and effective prophylactics and therapeutics against infection of its causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as 2019 novel coronavirus (2019-nCoV). The CoV spike (S) protein plays the most important roles in viral attachment, fusion and entry, and serves as a target for development of antibodies, entry inhibitors and vaccines. Here, we identified the receptor-binding domain (RBD) in SARS-CoV-2 S protein and found that the RBD protein bound strongly to human and bat angiotensin-converting enzyme 2 (ACE2) receptors. SARS-CoV-2 RBD exhibited significantly higher binding affinity to ACE2 receptor than SARS-CoV RBD and could block the binding and, hence, attachment of SARS-CoV-2 RBD and SARS-CoV RBD to ACE2-expressing cells, thus inhibiting their infection to host cells. SARS-CoV RBD-specific antibodies could cross-react with SARS-CoV-2 RBD protein, and SARS-CoV RBD-induced antisera could cross-neutralize SARS-CoV-2, suggesting the potential to develop SARS-CoV RBD-based vaccines for prevention of SARS-CoV-2 and SARS-CoV infection.
- 41Sun, C.; Chen, L.; Yang, J. SARS-CoV-2 and SARS-CoV spike-RBD structure and receptor binding comparison and potential implications on neutralizing antibody and vaccine development. bioRxiv , February 20, 2020, ver. 1. DOI: 10.1101/2020.02.16.951723 .Google ScholarThere is no corresponding record for this reference.
- 42Xia, S.; Yan, L.; Xu, W.; Agrawal, A. S.; Algaissi, A.; Tseng, C. K.; Wang, Q.; Du, L.; Tan, W.; Wilson, I. A.; Jiang, S.; Yang, B.; Lu, L. A pan-coronavirus fusion inhibitor targeting the HR1mdomain of human coronavirus spike. Sci. Adv. 2019, 5, eaav4580 DOI: 10.1126/sciadv.aav4580Google ScholarThere is no corresponding record for this reference.
- 43Rathod, S. B.; Prajapati, P. B.; Punjabi, L. B.; Prajapati, K. N.; Chauhan, N.; Mansuri, M. F. Peptide modelling and screening against human ACE2 and spike glycoprotein RBD of SARS CoV 2. In Silico Pharmacol. 2020, 8, 3, DOI: 10.1007/s40203-020-00055-wGoogle Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s3kt1aqsQ%253D%253D&md5=41a5887a9cbbf2a0fa7e9cd70804aceaPeptide modelling and screening against human ACE2 and spike glycoprotein RBD of SARS-CoV-2Rathod Shravan B; Prajapati Pravin B; Prajapati Kuntal N; Punjabi Lata B; Chauhan Neha; Mansuri Mohmedyasin FIn silico pharmacology (2020), 8 (1), 3 ISSN:2193-9616.Outbreak of Coronavirus Disease 2019 (COVID-19) has become a great challenge for scientific community globally. Virus enters cell through spike glycoprotein fusion with ACE2 (Angiotensin-Converting Enzyme 2) human receptor. Hence, spike glycoprotein of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a potential target for diagnostics, vaccines, and antibodies. Also, virus entry can be prevented by blocking ACE2 thus, ACE2 can be considered potential target for therapeutics. As being highly specific, safe and efficacious, peptides hold their place in therapeutics. In present study, we retrieved sequence of 70 peptides from Antiviral Peptide Database (AVPdb), modelled them using 3D structure predicting web tool and docked them with receptor binding domain (RBD) of spike protein and human host receptor ACE2 using peptide-protein docking. It was observed that peptides have more affinity towards ACE2 in comparison with spike RBD. Interestingly it was noticed that most of the peptides bind to RBM (residue binding motif) which is responsible for ACE2 binding at the interface of RBD while, for ACE2, peptides prefer to bind the core cavity rather than RBD binding interface. To further investigate how peptides at the interface of RBD or ACE2 alter the binding between RBD and ACE2, protein-protein docking of RBD and ACE2 with and without peptides was performed. Peptides, AVP0671 at RBD and AVP1244 at ACE2 interfaces significantly reduce the binding affinity and change the orientation of RBD and ACE2 binding. This finding suggests that peptides can be used as a drug to inhibit virus entry in cells to stop COVID-19 pandemic in the future after experimental evidences.
- 44Watson, A.; Ferreira, L.; Hwang, P.; Xu, J.; Stroud, R. Peptide antidotes to SARS-CoV-2 (COVID-19). bioRxiv , August 6, 2020, ver. 1. DOI: 10.1101/2020.08.06.238915 .Google ScholarThere is no corresponding record for this reference.
- 45Singh, A.; Thakur, M.; Sharma, L. K.; Chandra, K. Designing a multi epitope peptide based vaccine against SARS CoV 2. Sci. Rep. 2020, 10, 16219, DOI: 10.1038/s41598-020-73371-yGoogle Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvF2ksbrM&md5=47ec0379065f691b7fed33b5216556c5Designing a multi-epitope peptide based vaccine against SARS-CoV-2Singh, Abhishek; Thakur, Mukesh; Sharma, Lalit Kumar; Chandra, KailashScientific Reports (2020), 10 (1), 16219CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)COVID-19 pandemic has resulted in 16,114,449 cases with 646,641 deaths from the 217 countries, or territories as on July 27th 2020. Due to multifaceted issues and challenges in the implementation of the safety and preventive measures, inconsistent coordination between societies-governments and most importantly lack of specific vaccine to SARS-CoV-2, the spread of the virus that initially emerged at Wuhan is still uprising after taking a heavy toll on human life. In the present study, we mapped immunogenic epitopes present on the four structural proteins of SARS-CoV-2 and we designed a multi-epitope peptide based vaccine that, demonstrated a high immunogenic response with a vast application on world's human population. On codon optimization and in-silico cloning, we found that candidate vaccine showed high expression in E. coli and immune simulation resulted in inducing a high level of both B-cell and T-cell mediated immunity. The results predicted that exposure of vaccine by administrating three injections significantly subsidized the antigen growth in the system. The proposed candidate vaccine found promising by yielding desired results and hence, should be validated by practical experimentations for its functioning and efficacy to neutralize SARS-CoV-2.
- 46Molina, R.; Oliva, B.; Fernandez-Fuentes, N. A collection of designed peptides to target SARS-Cov-2 – ACE2 interaction: PepI-Covid19 database. bioRxiv , April 29, 2020, ver. 1. DOI: 10.1101/2020.04.28.051789 .Google ScholarThere is no corresponding record for this reference.
- 47Zhao, H.; To, K. K. W.; Sze, K. H.; Yung, T. T.; Bian, M.; Lam, H.; Yeung, M. L.; Li, C.; Chu, H.; Yuen, K. Y. A broad-spectrum virus- and host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV-2. Nat. Commun. 2020, 11, 4252, DOI: 10.1038/s41467-020-17986-9Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs12rurbM&md5=9d48f2626060fca048216f3a6cfa840dA broad-spectrum virus- and host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV-2Zhao, Hanjun; To, Kelvin K. W.; Sze, Kong-Hung; Yung, Timothy Tin-Mong; Bian, Mingjie; Lam, Hoiyan; Yeung, Man Lung; Li, Cun; Chu, Hin; Yuen, Kwok-YungNature Communications (2020), 11 (1), 4252CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The 2019 novel respiratory virus (SARS-CoV-2) causes COVID-19 with rapid global socioeconomic disruptions and disease burden to healthcare. The COVID-19 and previous emerging virus outbreaks highlight the urgent need for broad-spectrum antivirals. A defensin-like peptide P9R exhibited potent antiviral activity against pH-dependent viruses that require endosomal acidification for virus infection, including the enveloped pandemic A(H1N1)pdm09 virus, avian influenza A(H7N9) virus, coronaviruses (SARS-CoV-2, MERS-CoV and SARS-CoV), and the non-enveloped rhinovirus. P9R can significantly protect mice from lethal challenge by A(H1N1)pdm09 virus and shows low possibility to cause drug-resistant virus. Mechanistic studies indicate that the antiviral activity of P9R depends on the direct binding to viruses and the inhibition of virus-host endosomal acidification, which provides a proof of concept that virus-binding alk. peptides can broadly inhibit pH-dependent viruses. These results suggest that the dual-functional virus- and host-targeting P9R can be a promising candidate for combating pH-dependent respiratory viruses.
- 48Han, D. P.; Penn-Nicholson, A.; Cho, M. W. Identification of critical determinants on ACE2 for SARS-CoV entry and development of a potent entry inhibitor. Virology 2006, 350 (1), 15– 25, DOI: 10.1016/j.virol.2006.01.029Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XlsFKrt74%253D&md5=1db622935cd76ac9a4a74cf6f6b186d5Identification of critical determinants on ACE2 for SARS-CoV entry and development of a potent entry inhibitorHan, Dong P.; Penn-Nicholson, Adam; Cho, Michael W.Virology (2006), 350 (1), 15-25CODEN: VIRLAX; ISSN:0042-6822. (Elsevier)Severe acute respiratory syndrome (SARS) is caused by a novel coronavirus, SARS-CoV. Virus entry into cells is mediated through interactions between spike (S) glycoprotein and angiotensin-converting enzyme 2 (ACE2). Alanine scanning mutagenesis anal. was performed to identify determinants on ACE2 crit. for SARS-CoV infection. Results indicated that charged amino acids between residues 22 and 57 were important, K26 and D30, in particular. Peptides representing various regions of ACE2 crit. for virus infection were chem. synthesized and evaluated for antiviral activity. Two peptides (a.a. 22-44 and 22-57) exhibited a modest antiviral activity with IC50 of about 50 μM and 6 μM, resp. One peptide comprised of two discontinuous segments of ACE2 (a.a. 22-44 and 351-357) artificially linked together by glycine, exhibited a potent antiviral activity with IC50 of about 0.1 μM. This novel peptide is a promising candidate as a therapeutic agent against this deadly emerging pathogen.
- 49Shang, J.; Ye, G.; Shi, K.; Wan, Y.; Luo, C.; Aihara, H.; Geng, Q.; Auerbach, A.; Li, F. Structural basis of receptor recognition by SARS-CoV-2. Nature 2020, 581, 221– 224, DOI: 10.1038/s41586-020-2179-yGoogle Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtbs%253D&md5=33bc9151641b2adcfb0dbf446621a1dcStructural basis of receptor recognition by SARS-CoV-2Shang, Jian; Ye, Gang; Shi, Ke; Wan, Yushun; Luo, Chuming; Aihara, Hideki; Geng, Qibin; Auerbach, Ashley; Li, FangNature (London, United Kingdom) (2020), 581 (7807), 221-224CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A novel severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) recently emerged and is rapidly spreading in humans, causing COVID-191,2. A key to tackling this pandemic is to understand the receptor recognition mechanism of the virus, which regulates its infectivity, pathogenesis and host range. SARS-CoV-2 and SARS-CoV recognize the same receptor-angiotensin-converting enzyme 2 (ACE2)-in humans3,4. Here we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 (engineered to facilitate crystn.) in complex with ACE2. In comparison with the SARS-CoV RBD, an ACE2-binding ridge in SARS-CoV-2 RBD has a more compact conformation; moreover, several residue changes in the SARS-CoV-2 RBD stabilize two virus-binding hotspots at the RBD-ACE2 interface. These structural features of SARS-CoV-2 RBD increase its ACE2-binding affinity. Addnl., we show that RaTG13, a bat coronavirus that is closely related to SARS-CoV-2, also uses human ACE2 as its receptor. The differences among SARS-CoV-2, SARS-CoV and RaTG13 in ACE2 recognition shed light on the potential animal-to-human transmission of SARS-CoV-2. This study provides guidance for intervention strategies that target receptor recognition by SARS-CoV-2.
- 50Zhou, T.; Tsybovsky, Y.; Gorman, J.; Rapp, M.; Cerutti, G. Cryo-EM structures of SARS-CoV-2 spike without and with ACE2 reveal a pH-dependent switch to mediate endosomal positioning of receptor-binding domains. Cell Host Microbe 2020, 28 (6), 867– 879, DOI: 10.1016/j.chom.2020.11.004Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisVagtrjF&md5=e2cdd111aa8d7ec2ec774c62c26e6710Cryo-EM Structures of SARS-CoV-2 Spike without and with ACE2 Reveal a pH-Dependent Switch to Mediate Endosomal Positioning of Receptor-Binding DomainsZhou, Tongqing; Tsybovsky, Yaroslav; Gorman, Jason; Rapp, Micah; Cerutti, Gabriele; Chuang, Gwo-Yu; Katsamba, Phinikoula S.; Sampson, Jared M.; Schon, Arne; Bimela, Jude; Boyington, Jeffrey C.; Nazzari, Alexandra; Olia, Adam S.; Shi, Wei; Sastry, Mallika; Stephens, Tyler; Stuckey, Jonathan; Teng, I-Ting; Wang, Pengfei; Wang, Shuishu; Zhang, Baoshan; Friesner, Richard A.; Ho, David D.; Mascola, John R.; Shapiro, Lawrence; Kwong, Peter D.Cell Host & Microbe (2020), 28 (6), 867-879.e5CODEN: CHMECB; ISSN:1931-3128. (Elsevier Inc.)The SARS-CoV-2 spike employs mobile receptor-binding domains (RBDs) to engage the human ACE2 receptor and to facilitate virus entry, which can occur through low-pH-endosomal pathways. To understand how ACE2 binding and low pH affect spike conformation, we detd. cryo-electron microscopy structures-at serol. and endosomal pH-delineating spike recognition of up to three ACE2 mols. RBDs freely adopted "up" conformations required for ACE2 interaction, primarily through RBD movement combined with smaller alterations in neighboring domains. In the absence of ACE2, single-RBD-up conformations dominated at pH 5.5, resolving into a solitary all-down conformation at lower pH. Notably, a pH-dependent refolding region (residues 824-858) at the spike-interdomain interface displayed dramatic structural rearrangements and mediated RBD positioning through coordinated movements of the entire trimer apex. These structures provide a foundation for understanding prefusion-spike mechanics governing endosomal entry; we suggest that the low pH all-down conformation potentially facilitates immune evasion from RBD-up binding antibody.
- 51Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; Wang, X. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020, 581, 215– 220, DOI: 10.1038/s41586-020-2180-5Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtL8%253D&md5=279c60143e8e5eb505457e0778baa8efStructure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptorLan, Jun; Ge, Jiwan; Yu, Jinfang; Shan, Sisi; Zhou, Huan; Fan, Shilong; Zhang, Qi; Shi, Xuanling; Wang, Qisheng; Zhang, Linqi; Wang, XinquanNature (London, United Kingdom) (2020), 581 (7807), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new and highly pathogenic coronavirus (severe acute respiratory syndrome coronavirus-2, SARS-CoV-2) caused an outbreak in Wuhan city, Hubei province, China, starting from Dec. 2019 that quickly spread nationwide and to other countries around the world1-3. Here, to better understand the initial step of infection at an at. level, we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2. The overall ACE2-binding mode of the SARS-CoV-2 RBD is nearly identical to that of the SARS-CoV RBD, which also uses ACE2 as the cell receptor4. Structural anal. identified residues in the SARS-CoV-2 RBD that are essential for ACE2 binding, the majority of which either are highly conserved or share similar side chain properties with those in the SARS-CoV RBD. Such similarity in structure and sequence strongly indicate convergent evolution between the SARS-CoV-2 and SARS-CoV RBDs for improved binding to ACE2, although SARS-CoV-2 does not cluster within SARS and SARS-related coronaviruses1-3,5. The epitopes of two SARS-CoV antibodies that target the RBD are also analyzed for binding to the SARS-CoV-2 RBD, providing insights into the future identification of cross-reactive antibodies.
- 52Sadremomtaz, A.; Ali, A. M.; Jouyandeh, F.; Balalaie, S.; Navari, R. Molecular docking, synthesis and biological evaluation of vascular endothelial growth factor (VEGF) B based peptide as antiangiogenic agent targeting the second domain of the vascular endothelial growth factor receptor 1 (VEGFR1D2) for anticancer application. Sig. Transduct. Target Ther. 2020, 5, 76, DOI: 10.1038/s41392-020-0177-zGoogle ScholarThere is no corresponding record for this reference.
- 53Sadremomtaz, A.; Mansouri, K.; Alemzadeh, G.; Safa, M.; Rastaghi, A. E.; Asghari, S. M. Dual blockade of VEGFR1 and VEGFR2 by a novel peptide abrogates VEGF driven angiogenesis, tumor growth, and metastasis through PI3K/ AKT and MAPK/ERK1/2 pathway. Biochim. Biophys. Acta, Gen. Subj. 2018, 1862, 2688– 2700, DOI: 10.1016/j.bbagen.2018.08.013Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs1Wnu7vJ&md5=ce96e6bf828c9f08f06949e6bdb3a295Dual blockade of VEGFR1 and VEGFR2 by a novel peptide abrogates VEGF-driven angiogenesis, tumor growth, and metastasis through PI3K/AKT and MAPK/ERK1/2 pathwaySadremomtaz, Afsaneh; Mansouri, Kamran; Alemzadeh, Golnaz; Safa, Majid; Rastaghi, Ahmadreza Esmaeili; Asghari, S. MohsenBiochimica et Biophysica Acta, General Subjects (2018), 1862 (12), 2688-2700CODEN: BBGSB3; ISSN:0304-4165. (Elsevier B.V.)Neutralization of vascular endothelial growth factor receptor 1 (VEGFR1) and/or VEGFR2 is a widely used means of inhibiting tumor angiogenesis. Based on the complex X-ray structures of VEGFA/VEGFR1, VEGFA/VEGFR2, and VEGFB/VEGFR1, a peptide (referred to as VGB) was designed to simultaneously bind to VEGFR1 and VEGFR2, and binding, antiangiogenic and antitumor properties of the peptide was investigated in vitro. VGB bound to both VEGFR1 and VEGFR2 in human umbilical vein endothelial cells (HUVECs), and inhibited the proliferation of HUVECs, 4 T1 mammary carcinoma tumor (MCT) cells in human umbilical vein endothelial cells (HUVECs) and 4T1 mammary carcinoma tumor (MCT) cells, and inhibited the proliferation of HUVE, 4T1 MCT, and U87 glioblastoma cells. Through abrogation of AKT and ERK1/2 phosphorylation, VEGFA-stimulated proliferation, migration, and two- and three-dimensional tube formation in HUVECs were inhibited more potently by VGB than by bevacizumab. In a murine 4 T1 MCT model, VGB strongly inhibited tumor growth without causing wt. loss, accompanied by inhibition of AKT and ERK1/2 phosphorylation, a significant decrease in tumor cell proliferation (Ki-67 expression), angiogenesis (CD31 and CD34 expression), an increase in apoptosis index (increased TUNEL staining and p53 expression and decreased Bcl-2 expression), and the suppression of systematic spreading of the tumor (reduced NF-κB and MMP-9 and increased E-cadherin expression). The dual specificity of VGB for VEGFR1 and VEGFR2, through which the PI3K/AKT and MAPK/ERK1/2 signaling pathways can be abrogated and, subsequently, angiogenesis, tumor growth, and metastasis are inhibited. This study demonstrated that simultaneous blockade of VEGFR1 and VEGFR2 downstream cascades is an effective means for treatment of various angiogenic disorders, esp. cancer.
- 54Sadremomtaz, A.; Kobarfard, F.; Mansouri, K.; Mirzanejad, L.; Asghari, S. M. Suppression of migratory and metastatic pathways via blocking VEGFR1 and VEGFR2. J. Recept. Signal Transduction Res. 2018, 38, 432– 441, DOI: 10.1080/10799893.2019.1567785Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cbpvFartg%253D%253D&md5=e67668413d5631e86f813c3d8d1baed2Suppression of migratory and metastatic pathways via blocking VEGFR1 and VEGFR2Sadremomtaz Afsaneh; Mirzanejad Laleh; Asghari S Mohsen; Kobarfard Farzad; Mansouri KamranJournal of receptor and signal transduction research (2018), 38 (5-6), 432-441 ISSN:.BACKGROUND: Vascular endothelial growth factor (VEGF) A and B are endothelial cell mitogens whose ligation to VEGFR1/VEGFR2 drives tumor angiogenesis and metastasis, and epithelial-mesenchymal transition (EMT). Blockade of these signaling axes could be obtained by disturbing the interactions between VEGFA and/or VEGFB with VEGFR1 and/or VEGFR2. METHODS: A 14-mer peptide (VGB) that recognizes both VEGFR1 and VEGFR2 were investigated for its inhibitory effects on the VEGF-induced proliferation and migration using MTT and scratch assay, respectively. Downstream signaling pathways were also assessed by quantitative estimation of gene and protein expression using real-time PCR and immunohistochemistry (IHC). RESULTS: We investigated the inhibitory effects of VGB on downstream mediators of metastasis, including epithelial-cadherin (E-cadherin), matrix metalloprotease-9 (MMP-9), cancer myelocytomatosis (c-Myc), and nuclear factor-κβ (NF-κβ), and migration, comprising focal adhesion kinase (FAK) and its substrate Paxilin. VGB inhibited the VEGF-induced proliferation of human umbilical vein endothelial cells (HUVECs), 4T1 and U87 cells in a time- and dose-dependent manner and migration of HUVECs. Based on IHC analyses, treatment of 4T1 mammary carcinoma tumor with VGB led to the suppression of p-AKT, p-ERK1/2, MMP-9, NF-κβ, and activation of E-cadherin compared with PBS-treated controls. Moreover, quantitative real-time PCR analyses of VGB-treated tumors revealed the reduced expression level of FAK, Paxilin, NF-κβ, MMP-9, c-Myc, and increased expression level of E-cadherin compared to PBS-treated controls. CONCLUSIONS: Our results demonstrated that simultaneous blockade of VEGFR1/VEGFR2 is an effective strategy to fight solid tumors by targeting a wider range of mediators involved in tumor angiogenesis, growth, and metastasis.
- 55Abraham, M. J.; Murtola, T.; Schulz, R.; Pall, S.; Smith, J. C.; Hess, B.; Lindahl, E. GROMACS: High performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX. 2015, 1 (2), 19– 25, DOI: 10.1016/j.softx.2015.06.001Google ScholarThere is no corresponding record for this reference.
- 56Jo, S.; Kim, T.; Iyer, V. G.; Im, W. CHARMM-GUI: a web-based graphical user interface for CHARMM. J. Comput. Chem. 2008, 29, 1859– 1865, DOI: 10.1002/jcc.20945Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXosVKksbc%253D&md5=112a3dd61d792b040f9f716b32220d7eCHARMM-GUI: a web-based graphical user interface for CHARMMJo, Sunhwan; Kim, Taehoon; Iyer, Vidyashankara G.; Im, WonpilJournal of Computational Chemistry (2008), 29 (11), 1859-1865CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)CHARMM is an academic research program used widely for macromol. mechanics and dynamics with versatile anal. and manipulation tools of at. coordinates and dynamics trajectories. CHARMM-GUI, http://www.charmm-gui.org, has been developed to provide a web-based graphical user interface to generate various input files and mol. systems to facilitate and standardize the usage of common and advanced simulation techniques in CHARMM. The web environment provides an ideal platform to build and validate a mol. model system in an interactive fashion such that, if a problem is found through visual inspection, one can go back to the previous setup and regenerate the whole system again. In this article, we describe the currently available functional modules of CHARMM-GUI Input Generator that form a basis for the advanced simulation techniques. Future directions of the CHARMM-GUI development project are also discussed briefly together with other features in the CHARMM-GUI website, such as Archive and Movie Gallery.
- 57Gowers, R. J.; Linke, M.; Barnoud, J.; Reddy, T. J. E.; Melo, M. N.; Seyler, S. MDanalysis: a python package for the rapid analysis of molecular dynamics simulations. Proceedings of the 15th python in science conference 2016, 98– 105, DOI: 10.25080/Majora-629e541a-00eGoogle ScholarThere is no corresponding record for this reference.
- 58Salentin, S.; Schreiber, S.; Haupt, V. J.; Adasme, M. F.; Schroeder, M. PLIP: fully automated protein–ligand interaction profiler. Nucleic Acids Res. 2015, 43, W443– W447, DOI: 10.1093/nar/gkv315Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVymtbrI&md5=4878881a7bd715e5585682876e102e65PLIP: fully automated protein-ligand interaction profilerSalentin, Sebastian; Schreiber, Sven; Haupt, V. Joachim; Adasme, Melissa F.; Schroeder, MichaelNucleic Acids Research (2015), 43 (W1), W443-W447CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The characterization of interactions in protein-ligand complexes is essential for research in structural bioinformatics, drug discovery and biol. However, comprehensive tools are not freely available to the research community. Here, we present the protein-ligand interaction profiler (PLIP), a novel web service for fully automated detection and visualization of relevant non-covalent protein-ligand contacts in 3D structures, freely available at projects.biotec.tu-dresden.de/plip-web. The input is either a Protein Data Bank structure, a protein or ligand name, or a custom protein-ligand complex (e.g. from docking). In contrast to other tools, the rule-based PLIP algorithm does not require any structure prepn. It returns a list of detected interactions on single atom level, covering seven interaction types (hydrogen bonds, hydrophobic contacts, pi-stacking, pi-cation interactions, salt bridges, water bridges and halogen bonds). PLIP stands out by offering publication-ready images, PyMOL sesions files to generate custom images and parsable result files to facilitate successive data processing. The full python source code is available for download on the website. PLIP's command-line mode allows for high-throughput interaction profiling.
- 59Kayikci, M.; Venkatakrishnan, A. J.; Scott-Brown, J. Visualization and analysis of non-covalent contacts using the protein contacts atlas. Nat. Struct. Mol. Biol. 2018, 25, 185– 194, DOI: 10.1038/s41594-017-0019-zGoogle Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlCqu7rF&md5=b72bfcbc3e4e573920fb9f1f0403de88Visualization and analysis of non-covalent contacts using the Protein Contacts AtlasKayikci, Melis; Venkatakrishnan, A. J.; Scott-Brown, James; Ravarani, Charles N. J.; Flock, Tilman; Babu, M. MadanNature Structural & Molecular Biology (2018), 25 (2), 185-194CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)Visualizations of biomol. structures empower us to gain insights into biol. functions, generate testable hypotheses, and communicate biol. concepts. Typical visualizations (such as ball and stick) primarily depict covalent bonds. In contrast, non-covalent contacts between atoms, which govern normal physiol., pathogenesis, and drug action, are seldom visualized. We present the Protein Contacts Atlas, an interactive resource of non-covalent contacts from over 100,000 PDB crystal structures. We developed multiple representations for visualization and anal. of non-covalent contacts at different scales of organization: atoms, residues, secondary structure, subunits, and entire complexes. The Protein Contacts Atlas enables researchers from different disciplines to investigate diverse questions in the framework of non-covalent contacts, including the interpretation of allostery, disease mutations and polymorphisms, by exploring individual subunits, interfaces, and protein-ligand contacts and by mapping external information. The Protein Contacts Atlas is available at http://www.mrc-lmb.cam.ac.uk/pca/ and also through PDBe.
- 60Azad, T.; Singaravelu, R.; Taha, Z.; Boulton, S. Nanoluciferase complementation-based biosensor reveals the importance of N- linked glycosylation of SARS-CoV-2 spike for viral entry. Mol. Ther. 2021, 29, 1984, DOI: 10.1016/j.ymthe.2021.02.007Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXltV2hsr0%253D&md5=f96a869c49349aef8a2946ee1d0e5090Nanoluciferase complementation-based bioreporter reveals the importance of N-linked glycosylation of SARS-CoV-2 S for viral entryAzad, Taha; Singaravelu, Ragunath; Taha, Zaid; Jamieson, Taylor R.; Boulton, Stephen; Crupi, Mathieu J. F.; Martin, Nikolas T.; Brown, Emily E. F.; Poutou, Joanna; Ghahremani, Mina; Pelin, Adrian; Nouri, Kazem; Rezaei, Reza; Marshall, Christopher Boyd; Enomoto, Masahiro; Arulanandam, Rozanne; Alluqmani, Nouf; Samson, Reuben; Gingras, Anne-Claude; Cameron, D. William; Greer, Peter A.; Ilkow, Carolina S.; Diallo, Jean-Simon; Bell, John C.Molecular Therapy (2021), 29 (6), 1984-2000CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)The ongoing COVID-19 pandemic has highlighted the immediate need for the development of antiviral therapeutics targeting different stages of the SARS-CoV-2 life cycle. We developed a bioluminescence-based bioreporter to interrogate the interaction between the SARS-CoV-2 viral spike (S) protein and its host entry receptor, angiotensin-converting enzyme 2 (ACE2). The bioreporter assay is based on a nanoluciferase complementation reporter, composed of two subunits, large BiT and small BiT, fused to the S receptor-binding domain (RBD) of the SARS-CoV-2 S protein and ACE2 ectodomain, resp. Using this bioreporter, we uncovered crit. host and viral determinants of the interaction, including a role for glycosylation of asparagine residues within the RBD in mediating successful viral entry. We also demonstrate the importance of N-linked glycosylation to the RBD's antigenicity and immunogenicity. Our study demonstrates the versatility of our bioreporter in mapping key residues mediating viral entry as well as screening inhibitors of the ACE2-RBD interaction. Our findings point toward targeting RBD glycosylation for therapeutic and vaccine strategies against SARS-CoV-2.
- 61Spiliotopoulos, D.; Kastritis, P. L.; Melquiond, A. S.; Bonvin, A. M.; Musco, G.; Rocchia, W.; Spitaleri, A. dMM-PBSA: A new HADDOCK scoring function for protein-peptide docking. Front Mol. Biosci. 2016, 3, 46, DOI: 10.3389/fmolb.2016.00046Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvFSitL%252FO&md5=01d6e4223bf1a6329e25ec6ad7802cd0dMM-PBSA: a new HADDOCK scoring function for protein-peptide dockingSpiliotopoulos, Dimitrios; Kastritis, Panagiotis L.; Melquiond, Adrien S. J.; Bonvin, Alexandre M. J. J.; Musco, Giovanna; Rocchia, Walter; Spitaleri, AndreaFrontiers in Molecular Biosciences (2016), 3 (), 46/1-46/13CODEN: FMBRBS; ISSN:2296-889X. (Frontiers Media S.A.)Mol.-docking programs coupled with suitable scoring functions are now established and very useful tools enabling computational chemists to rapidly screen large chem. databases and thereby to identify promising candidate compds. for further exptl. processing. In a broader scenario, predicting binding affinity is one of the most crit. and challenging components of computer-aided structure-based drug design. The development of a mol. docking scoring function which in principle could combine both features, namely ranking putative poses and predicting complex affinity, would be of paramount importance. Here, we systematically investigated the performance of the MM-PBSA approach, using two different Poisson-Boltzmann solvers (APBS and DelPhi), in the currently rising field of protein-peptide interactions (PPIs), identifying the correct binding conformations of 19 different protein-peptide complexes and predicting their binding free energies. First, we scored the decoy structures from HADDOCK calcn. via the MM-PBSA approach in order to assess the capability of retrieving near-native poses in the best-scoring clusters and of evaluating the corresponding free energies of binding. MM-PBSA behaves well in finding the poses corresponding to the lowest binding free energy. In order to improve the MM-PBSA-based scoring function, we dampened the MM-PBSA solvation and coulombic terms by 0.2, as proposed in the HADDOCK score and LIE approaches.
- 62Li, W.; Zhang, C.; Sui, J.; Kuhn, J. H.; Moore, M. J.; Luo, S.; Wong, S. K.; Huang, I. C.; Xu, K.; Vasilieva, N.; Murakami, A.; He, Y.; Marasco, W. A.; Guan, Y.; Choe, H.; Farzan, M. Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2. EMBO J. 2005, 24, 1634– 1643, DOI: 10.1038/sj.emboj.7600640Google Scholar100https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXktFequ7o%253D&md5=7b7d477b5376c04a7607f507b63702f2Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2Li, Wenhui; Zhang, Chengsheng; Sui, Jianhua; Kuhn, Jens H.; Moore, Michael J.; Luo, Shiwen; Wong, Swee-Kee; Huang, I-Chueh; Xu, Keming; Vasilieva, Natalya; Murakami, Akikazu; He, Yaqing; Marasco, Wayne A.; Guan, Yi; Choe, Hyeryun; Farzan, MichaelEMBO Journal (2005), 24 (8), 1634-1643CODEN: EMJODG; ISSN:0261-4189. (Nature Publishing Group)Human angiotensin-converting enzyme 2 (ACE2) is a functional receptor for SARS coronavirus (SARS-CoV). Here we identify the SARS-CoV spike (S)-protein-binding site on ACE2. We also compare S proteins of SARS-CoV isolated during the 2002-2003 SARS outbreak and during the much less severe 2003-2004 outbreak, and from palm civets, a possible source of SARS-CoV found in humans. All three S proteins bound to and utilized palm-civet ACE2 efficiently, but the latter two S proteins utilized human ACE2 markedly less efficiently than did the S protein obtained during the earlier human outbreak. The lower affinity of these S proteins could be complemented by altering specific residues within the S-protein-binding site of human ACE2 to those of civet ACE2, or by altering S-protein residues 479 and 487 to residues conserved during the 2002-2003 outbreak. Collectively, these data describe mol. interactions important to the adaptation of SARS-CoV to human cells, and provide insight into the severity of the 2002-2003 SARS epidemic.
- 63Elbe, S.; Buckland-Merrett, G. Data, disease and diplomacy: GISAID’s innovative contribution to global health. Global Challenges 2017, 1, 33, DOI: 10.1002/gch2.1018Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MnjtFKmug%253D%253D&md5=ec5a87c7612a5922dd7bd76718359f20Data, disease and diplomacy: GISAID's innovative contribution to global healthElbe Stefan; Buckland-Merrett GemmaGlobal challenges (Hoboken, NJ) (2017), 1 (1), 33-46 ISSN:.The international sharing of virus data is critical for protecting populations against lethal infectious disease outbreaks. Scientists must rapidly share information to assess the nature of the threat and develop new medical countermeasures. Governments need the data to trace the extent of the outbreak, initiate public health responses, and coordinate access to medicines and vaccines. Recent outbreaks suggest, however, that the sharing of such data cannot be taken for granted - making the timely international exchange of virus data a vital global challenge. This article undertakes the first analysis of the Global Initiative on Sharing All Influenza Data as an innovative policy effort to promote the international sharing of genetic and associated influenza virus data. Based on more than 20 semi-structured interviews conducted with key informants in the international community, coupled with analysis of a wide range of primary and secondary sources, the article finds that the Global Initiative on Sharing All Influenza Data contributes to global health in at least five ways: (1) collating the most complete repository of high-quality influenza data in the world; (2) facilitating the rapid sharing of potentially pandemic virus information during recent outbreaks; (3) supporting the World Health Organization's biannual seasonal flu vaccine strain selection process; (4) developing informal mechanisms for conflict resolution around the sharing of virus data; and (5) building greater trust with several countries key to global pandemic preparedness.
- 64Baskaran, K.; Duarte, J. M.; Biyani, N.; Bliven, S.; Capitani, G. A PDB-wide, evolution-based assessment of protein-protein interfaces. BMC Struct. Biol. 2014, 14, 1422, DOI: 10.1186/s12900-014-0022-0Google ScholarThere is no corresponding record for this reference.
- 65Amaral, M.; Kokh, D. B.; Bomke, J.; Wegener, A.; Buchstaller, H. P.; Eggenweiler, H. M. Protein conformational flexibility modulates kinetics and thermodynamics of drug binding. Nat. Commun. 2017, 8, 2276, DOI: 10.1038/s41467-017-02258-wGoogle Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mzksleqsg%253D%253D&md5=2ccf76a6569363af27b6f27cb1be5b7aProtein conformational flexibility modulates kinetics and thermodynamics of drug bindingAmaral M; Matias P; Amaral M; Wegener A; Frech M; Amaral M; Kokh D B; Wade R C; Bomke J; Buchstaller H P; Eggenweiler H M; Matias P; Sirrenberg C; Wade R C; Wade R CNature communications (2017), 8 (1), 2276 ISSN:.Structure-based drug design has often been restricted by the rather static picture of protein-ligand complexes presented by crystal structures, despite the widely accepted importance of protein flexibility in biomolecular recognition. Here we report a detailed experimental and computational study of the drug target, human heat shock protein 90, to explore the contribution of protein dynamics to the binding thermodynamics and kinetics of drug-like compounds. We observe that their binding properties depend on whether the protein has a loop or a helical conformation in the binding site of the ligand-bound state. Compounds bound to the helical conformation display slow association and dissociation rates, high-affinity and high cellular efficacy, and predominantly entropically driven binding. An important entropic contribution comes from the greater flexibility of the helical relative to the loop conformation in the ligand-bound state. This unusual mechanism suggests increasing target flexibility in the bound state by ligand design as a new strategy for drug discovery.
- 66Ferina, J.; Daggett, V. Visualizing protein folding and unfolding. J. Mol. Biol. 2019, 431, 1540– 1564, DOI: 10.1016/j.jmb.2019.02.026Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXksFKnuro%253D&md5=4296169975d3fdd59d279bb7afd26303Visualizing Protein Folding and UnfoldingFerina, Jennifer; Daggett, ValerieJournal of Molecular Biology (2019), 431 (8), 1540-1564CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. Protein folding/unfolding is a complicated process that defies high-resoln. characterization by exptl. methods. As an alternative, atomistic mol. dynamics simulations are now routinely employed to elucidate and magnify the accompanying conformational changes and the role of solvent in the folding process. However, the level of detail necessary to map the process at high spatial-temporal resoln. provides an overwhelming amt. of data. As more and better tools are developed for anal. of these large data sets and validation of the simulations, one is still left with the problem of visualizing the results in ways that provide insight into the folding/unfolding process. While viewing and interrogating static crystal structures has become commonplace, more and different approaches are required for dynamic, interconverting, unfolding, and refolding proteins. Here the authors review a variety of approaches, ranging from straightforward to complex and unintuitive for multiscale anal. and visualization of protein folding and unfolding.
- 67Zambrano, R.; Jamroz, M.; Szczasiuk, A.; Pujols, J.; Kmiecik, S.; Ventura, S. AGGRESCAN3D (A3D): server for prediction of aggregation properties of protein structures. Nucleic Acids Res. 2015, 43, W306– 313, DOI: 10.1093/nar/gkv359Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVymtbjK&md5=4d5a4d94fa0bf2744250860780e2a203AGGRESCAN3D (A3D): server for prediction of aggregation properties of protein structuresZambrano, Rafael; Jamroz, Michal; Szczasiuk, Agata; Pujols, Jordi; Kmiecik, Sebastian; Ventura, SalvadorNucleic Acids Research (2015), 43 (W1), W306-W313CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Protein aggregation underlies an increasing no. of disorders and constitutes a major bottleneck in the development of therapeutic proteins. Our present understanding on the mol. determinants of protein aggregation has crystd. in a series of predictive algorithms to identify aggregation-prone sites. A majority of these methods rely only on sequence. Therefore, they find difficulties to predict the aggregation properties of folded globular proteins, where aggregation-prone sites are often not contiguous in sequence or buried inside the native structure. The AGGRESCAN3D (A3D) server overcomes these limitations by taking into account the protein structure and the exptl. aggregation propensity scale from the well-established AGGRESCAN method. Using the A3D server, the identified aggregation-prone residues can be virtually mutated to design variants with increased soly., or to test the impact of pathogenic mutations. Addnl., A3D server enables to take into account the dynamic fluctuations of protein structure in soln., which may influence aggregation propensity. This is possible in A3D Dynamic Mode that exploits the CABS-flex approach for the fast simulations of flexibility of globular proteins.
- 68Goel, B.; Bhardwaj, N.; Tripathi, N.; Jain, S. K. Drug discovery of small molecules for the treatment of COVID-19: a review on clinical studies. Mini-Rev. Med. Chem. 2021, 21, 1431, DOI: 10.2174/1389557521666201228145755Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1Sjt7vP&md5=05d78cb3bc1278ab6ec389f4c403a0f8Drug Discovery of Small Molecules for the Treatment of COVID-19: A Review on Clinical StudiesGoel, Bharat; Bhardwaj, Nivedita; Tripathi, Nancy; Jain, Shreyans K.Mini-Reviews in Medicinal Chemistry (2021), 21 (12), 1431-1456CODEN: MMCIAE; ISSN:1389-5575. (Bentham Science Publishers Ltd.)A review. Recently, a sudden outbreak of novel coronavirus disease (COVID-19) was caused by a zoonotic virus known as severe acute respiratory syndrome coronavirus (SARS-CoV-2). It has caused pandemic situations around the globe affecting the lives of millions of people. So far, no drug has been approved for the treatment of SARS-CoV-2 infected patients. As of now, more than 1000 clin. trials are going on for repurposing of FDA-approved drugs and for evaluating the safety and efficiency of exptl. antiviral mols. to combat COVID-19. Since the development of new drugs may require months to years to reach the market, this review focusses on the potential of existing small mol. FDA approved drugs and the mols. already in the clin. pipeline against viral infections like HIV, hepatitis B, Ebola virus, and other viruses of coronavirus family (SARS-CoV and MERS-CoV). The review also discusses the natural products and traditional medicines in clin. studies against COVID-19. Currently, 1978 studies are active, 143 completed and 4 posted results (as of June 13, 2020) on clinicaltrials.gov.
- 69Pant, S.; Singh, M.; Ravichandiran, V.; Murty, U. S. N.; Srivastava, H. K. Peptide-like and small-molecule inhibitors against COVID-19. J. Biomol. Struct. Dyn. 2021, 39, 2904, DOI: 10.1080/07391102.2020.1757510Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVShurrO&md5=91f4cbaeddbc425f807f2ee413023d2cPeptide-like and small-molecule inhibitors against Covid-19Pant, Suyash; Singh, Meenakshi; Ravichandiran, V.; Murty, U. S. N.; Srivastava, Hemant KumarJournal of Biomolecular Structure and Dynamics (2021), 39 (8), 2904-2913CODEN: JBSDD6; ISSN:0739-1102. (Taylor & Francis Ltd.)Coronavirus disease strain (SARS-CoV-2) was discovered in 2019, and it is spreading very fast around the world causing the disease Covid-19. Currently, >1.6 million individuals are infected, and several thousand are dead across the globe because of Covid-19. We utilized the in-silico approaches to identify possible protease inhibitors against SARS-CoV-2. Potential compds. were screened from the CHEMBL database, ZINC database, FDA approved drugs, and mols. under clin. trials. Our study is based on 6Y2F and 6W63 co-crystd. structures available in the protein data bank (PDB). Seven hundred compds. from ZINC/CHEMBL databases and 1400 compds. from drug-bank were selected based on pos. interactions with the reported binding site. All the selected compds. were subjected to std.-precision (SP) and extra-precision (XP) mode of docking. Generated docked poses were carefully visualized for known interactions within the binding site. Mol. mechanics-generalized born surface area (MM-GBSA) calcns. were performed to screen the best compds. based on docking scores and binding energy values. Mol. dynamics (MD) simulations were carried out on 4 selected compds. from the CHEMBL database to validate the stability and interactions. MD simulations were also performed on the PDB structure 6YF2F to understand the differences between screened mols. and co-crystd. ligand. We screened 300 potential compds. from various databases, and 66 potential compds. from FDA approved drugs. Cobicistat, ritonavir, lopinavir, and darunavir are in the top screened mols. from FDA approved drugs. The screened drugs and mols. may be helpful in fighting with SARS-CoV-2 after further studies.
- 70Di Natale, C.; La Manna, S.; De Benedictis, I.; Brandi, P.; Marasco, D. Perspectives in peptide-based vaccination strategies for syndrome coronavirus 2 pandemic. Front. Pharmacol. 2020, 11, 578382, DOI: 10.3389/fphar.2020.578382Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsl2jsLg%253D&md5=c1b4fea4454c0207169e74a870617bbfPerspectives in peptide-based vaccination strategies for syndrome coronavirus 2 pandemicDi Natale, Concetta; La Manna, Sara; De Benedictis, Ilaria; Brandi, Paola; Marasco, DanielaFrontiers in Pharmacology (2020), 11 (), 578382CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)At the end of Dec. 2019, an epidemic form of respiratory tract infection now named COVID-19 emerged in Wuhan, China. It is caused by a newly identified viral pathogen, the severe acute respiratory syndrome coronavirus (SARS-CoV-2), which can cause severe pneumonia and acute respiratory distress syndrome. On Jan. 30, 2020, due to the rapid spread of infection, COVID-19 was declared as a global health emergency by the World Health Organization. Coronaviruses are enveloped RNA viruses belonging to the family of Coronaviridae, which are able to infect birds, humans and other mammals. The majority of human coronavirus infections are mild although already in 2003 and in 2012, the epidemics of SARS-CoV and Middle East Respiratory Syndrome coronavirus (MERSCoV), resp., were characterized by a high mortality rate. In this regard, many efforts have been made to develop therapeutic strategies against human CoV infections but, unfortunately, drug candidates have shown efficacy only into in vitro studies, limiting their use against COVID-19 infection. Actually, no treatment has been approved in humans against SARS-CoV-2, and therefore there is an urgent need of a suitable vaccine to tackle this health issue. However, the puzzled scenario of biol. features of the virus and its interaction with human immune response, represent a challenge for vaccine development. As expected, in hundreds of research labs. there is a running out of breath to explore different strategies to obtain a safe and quickly spreadable vaccine; and among others, the peptide-based approach represents a turning point as peptides have demonstrated unique features of selectivity and specificity toward specific targets. Peptide-based vaccines imply the identification of different epitopes both on human cells and virus capsid and the design of peptide/peptidomimetics able to counteract the primary host-pathogen interaction, in order to induce a specific host immune response. SARS-CoV-2 immunogenic regions are mainly distributed, as well as for other coronaviruses, across structural areas such as spike, envelope, membrane or nucleocapsid proteins. Herein, we aim to highlight the mol. basis of the infection and recent peptide-based vaccines strategies to fight the COVID-19 pandemic including their delivery systems.
- 71Pessi, A.; Bixler, S. L.; Soloveva, V.; Radoshitzky, S.; Retterer, C. Cholesterol-conjugated stapled peptides inhibit Ebola and Marburg viruses in vitro and in vivo. Antiviral Res. 2019, 171, 104592, DOI: 10.1016/j.antiviral.2019.104592Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVSgs7fO&md5=a5ca9bbc605a0e0618473ce5e7a3728fCholesterol-conjugated stapled peptides inhibit Ebola and Marburg viruses in vitro and in vivoPessi, Antonello; Bixler, Sandra L.; Soloveva, Veronica; Radoshitzky, Sheli; Retterer, Cary; Kenny, Tara; Zamani, Rouzbeh; Gomba, Glenn; Gharabeih, Dima; Wells, Jay; Wetzel, Kelly S.; Warren, Travis K.; Donnelly, Ginger; Van Tongeren, Sean A.; Steffens, Jesse; Duplantier, Allen J.; Kane, Christopher D.; Vicat, Pascale; Couturier, Valerie; Kester, Kent E.; Shiver, John; Carter, Kara; Bavari, SinaAntiviral Research (2019), 171 (), 104592CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)Currently, there are no therapeutics approved and the need for Ebola-specific therapeutics remains a gap. In search for anti-Ebola therapies we tested the idea of using inhibitory properties of peptides corresponding to the C-terminal heptad-repeat (HR2) domains of class I fusion proteins against EBOV infection. The fusion protein GP2 of EBOV belongs to class I, suggesting that a similar strategy to HIV may be applied to inhibit EBOV infection. The serum half-life of peptides was expanded by cholesterol conjugation to allow daily dosing. The peptides were further constrained to stabilize a helical structure to increase the potency of inhibition. The EC50s of lead peptides were in low micromolar range, as detd. by a high-content imaging test of EBOV-infected cells. Lead peptides were tested in an EBOV lethal mouse model and efficacy of the peptides were detd. following twice-daily administration of peptides for 9 days. The most potent peptide was able to protect mice from lethal challenge of mouse-adapted Ebola virus. These data show that engineered peptides coupled with cholesterol can inhibit viral prodn., protect mice against lethal EBOV infection, and may be used to build novel therapeutics against EBOV.
- 72Li, C. G.; Tang, W.; Chi, X. J.; Dong, Z. M.; Wang, X. X.; Wang, X. J. A cholesterol tag at the N terminus of the relatively broad-spectrum fusion inhibitory peptide targets an earlier stage of fusion glycoprotein activation and increases the peptide’s antiviral potency in vivo. J. Virol. 2013, 87, 9223– 9232, DOI: 10.1128/JVI.01153-13Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1WgtLvP&md5=d418cb40ba11ea15d7c50ad61ee2caaaA cholesterol tag at the N terminus of the relatively broad-spectrum fusion inhibitory peptide targets an earlier stage of fusion glycoprotein activation and increases the peptide's antiviral potency in vivoLi, Chuan-Gen; Wang, Tang; Chi, Xiao-Jing; Dong, Zhi-Ming; Wang, Xi-Xi; Wang, Xiao-JiaJournal of Virology (2013), 87 (16), 9223-9232CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)In previous work, we designed peptides that showed potent inhibition of Newcastle disease virus (NDV) and infectious bronchitis virus (IBV) infections in chicken embryos. In this study, we demonstrate that peptides modified with cholesterol or 3 U of polyethylene glycol (PEG3) conjugated to the peptides' N termini showed even more promising antiviral activities when tested in animal models. Both cholesterol- and cholesterol-PEG3-tagged peptides were able to protect chicken embryos from infection with different serotypes of NDV and IBV when administered 12 h prior to virus inoculation. In comparison, the untagged peptides required intervention closer to the time of viral inoculation to achieve a similar level of protection. I.m. injection of cholesterol-tagged peptide at 1.6 mg/kg 1 day before virus infection and then three times at 3-day intervals after viral inoculation protected 70% of the chickens from NDV infection. We further demonstrate that the cholesterol-tagged peptide has an in vivo half-life greater than that of untagged peptides. It also has the potential to cross the blood-brain barrier to enter the avian central nervous system (CNS). Finally, we show that the cholesterol-tagged peptide could play a role before the viral fusion peptide's insertion into the host cell and thereby target an earlier stage of fusion glycoprotein activation. Our findings are of importance for the further development of antivirals with broad-spectrum protective effects.
- 73Pessi, A. Cholesterol-conjugated peptide antivirals: a path to a rapid response to emerging viral diseases. J. Pept. Sci. 2015, 21, 379– 386, DOI: 10.1002/psc.2706Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntFCkt7w%253D&md5=56d038b929227c81bdc5e85cc562de75Cholesterol-conjugated peptide antivirals: a path to a rapid response to emerging viral diseasesPessi, AntonelloJournal of Peptide Science (2015), 21 (5), 379-386CODEN: JPSIEI; ISSN:1075-2617. (John Wiley & Sons Ltd.)A review. While it is now possible to identify and genetically fingerprint the causative agents of emerging viral diseases, often with extraordinary speed, suitable therapies cannot be developed with equiv. speed, because drug discovery requires information that goes beyond knowledge of the viral genome. Peptides, however, may represent a special opportunity. For all enveloped viruses, fusion between the viral and the target cell membrane is an obligatory step of the life cycle. Class I fusion proteins harbor regions with a repeating pattern of amino acids, the heptad repeats (HRs), that play a key role in fusion, and HR-derived peptides such as enfuvirtide, in clin. use for HIV, can block the process. Because of their characteristic sequence pattern, HRs are easily identified in the genome by means of computer programs, providing the sequence of candidate peptide inhibitors directly from genomic information. Moreover, a simple chem. modification, the attachment of a cholesterol group, can dramatically increase the antiviral potency of HR-derived inhibitors and simultaneously improve their pharmacokinetics. Further enhancement can be provided by dimerization of the cholesterol-conjugated peptide. The examples reported so far include inhibitors of retroviruses, paramyxoviruses, orthomyxoviruses, henipaviruses, coronaviruses, and filoviruses. For some of these viruses, in vivo efficacy has been demonstrated in suitable animal models. The combination of bioinformatic lead identification and potency/pharmacokinetics improvement provided by cholesterol conjugation may form the basis for a rapid response strategy, where development of an emergency cholesterol-conjugated therapeutic would immediately follow the availability of the genetic information of a new enveloped virus. Copyright © 2014 European Peptide Society and John Wiley & Sons, Ltd.
- 74Pettersen, E. F.; Goddard, T. D.; Huang, C. C.; Couch, G. S.; Greenblatt, D. M.; Meng, E. C.; Ferrin, T. E. UCSF chimera--a visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605– 1612, DOI: 10.1002/jcc.20084Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmvVOhsbs%253D&md5=944b175f440c1ff323705987cf937ee7UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 75Caporale, A.; Doti, N.; Monti, A.; Sandomenico, A.; Ruvo, M. Automatic procedures for the synthesis of difficult peptides using oxyma as activating reagent: A comparative study on the use of bases and on different deprotection and agitation conditions. Peptides 2018, 102, 38– 46, DOI: 10.1016/j.peptides.2018.02.006Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvVClsbw%253D&md5=e62fa59f680ab328407d6992db13b802Automatic procedures for the synthesis of difficult peptides using oxyma as activating reagent: A comparative study on the use of bases and on different deprotection and agitation conditionsCaporale, A.; Doti, N.; Monti, A.; Sandomenico, A.; Ruvo, M.Peptides (New York, NY, United States) (2018), 102 (), 38-46CODEN: PPTDD5; ISSN:0196-9781. (Elsevier)Solid-Phase Peptide Synthesis (SPPS) is a rapid and efficient methodol. for the chem. synthesis of peptides and small proteins. However, the assembly of peptide sequences classified as "difficult" poses severe synthetic problems in SPPS for the occurrence of extensive aggregation of growing peptide chains which often leads to synthesis failure. In this framework, we have investigated the impact of different synthetic procedures on the yield and final purity of three well-known "difficult peptides" prepd. using oxyma as additive for the coupling steps. In particular, we have comparatively investigated the use of piperidine and morpholine/DBU as deprotection reagents, the addn. of DIPEA, collidine and N-methylmorpholine as bases to the coupling reagent. Moreover, the effect of different agitation modalities during the acylation reactions has been investigated. Data obtained represent a step forward in optimizing strategies for the synthesis of "difficult peptides".
- 76Grimsley, G. R.; Pace, C. N. Spectrophotometric Determination of Protein Concentration. Curr. Protoc. Protein Sci. 2003, 3.1.1– 3.1.9, DOI: 10.1002/0471140864.ps0301s33Google ScholarThere is no corresponding record for this reference.
- 77Scopes, R.K. Measurement of protein by spectrophotometry at 205 nm. Anal. Biochem. 1974, 59, 277– 282, DOI: 10.1016/0003-2697(74)90034-7Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE2cXksVOjsb0%253D&md5=ff53a6ad935bde82fc8c8a60bb606760Measurement of protein by spectrophotometry at 205nmScopes, R. K.Analytical Biochemistry (1974), 59 (1), 277-82CODEN: ANBCA2; ISSN:0003-2697.A method was described for the measurement of protein concn. by using the peptide bond absorption at 205 nm. The extinction coeff. (ε) at 205 nm was estd., allowing for the absorption due to tryptophan and tyrosine residues, by measuring the absorbance at 280 nm as well as at 205 nm. The estd. ε205 was compared with the actual ε205 for a no. of proteins, the mean error being <2%. This was about 3 times better than using an av. ε2051mg/ml of 31 and approached the range of exptl. error inherent in any method of protein estn.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 25 publications.
- Lalita Mohan Behera, Pulkit Kr. Gupta, Manaswini Ghosh, Sucharita Shadangi, Soumendra Rana. A Rationally Designed Synthetic Antiviral Peptide Binder Targeting the Receptor-Binding Domain of SARS-CoV-2. The Journal of Physical Chemistry B 2024, 128
(19)
, 4631-4645. https://doi.org/10.1021/acs.jpcb.4c00241
- Angel J. Ruiz-Moreno, Raziel Cedillo-González, Luis Cordova-Bahena, Zhiqiang An, José L. Medina-Franco, Marco A. Velasco-Velázquez. Consensus Pharmacophore Strategy For Identifying Novel SARS-Cov-2 Mpro Inhibitors from Large Chemical Libraries. Journal of Chemical Information and Modeling 2024, 64
(6)
, 1984-1995. https://doi.org/10.1021/acs.jcim.3c01439
- Michael Quagliata, Maria Alfreda Stincarelli, Anna Maria Papini, Simone Giannecchini, Paolo Rovero. Antiviral Activity against SARS-CoV-2 of Conformationally Constrained Helical Peptides Derived from Angiotensin-Converting Enzyme 2. ACS Omega 2023, 8
(25)
, 22665-22672. https://doi.org/10.1021/acsomega.3c01436
- 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
- Mandy Schwarze, Alexandra Brakel, Ralf Hoffmann, Andor Krizsan. Peptides Corresponding to the Receptor‐Binding Domain (RBD) of Several SARS‐CoV‐2 Variants Of Concern Prevent Recognition of the Human ACE2 Receptor and Consecutive Cell Infections. ChemMedChem 2025, https://doi.org/10.1002/cmdc.202400973
- Nisha Bansal, Deepika Kathuria, Arockia M. Babu, Sonia Dhiman, Sorabh Lakhanpal, K. Nagendra Prasad, Roshan Kumar, Yogita Tyagi, Bhupinder Kumar, Mahendra Pratap Singh, Abhay M. Gaidhane. A perspective on small molecules targeting the renin–angiotensin–aldosterone system and their utility in cardiovascular diseases: exploring the structural insights for rational drug discovery and development. RSC Medicinal Chemistry 2025, 94 https://doi.org/10.1039/D4MD00720D
- Haixiang Ling, Yunhe Yuan, Fuli Chen, Jiuchuan Guo, Jinhong Guo, Diangeng Li. Thermophoretic Assay for Biosensing. Advanced Materials Technologies 2024, 9
(13)
https://doi.org/10.1002/admt.202301746
- Hongying Li, Jianrui Li, Jiayu Li, Hu Li, Xuekai Wang, Jing Jiang, Lei Lei, Han Sun, Mei Tang, Biao Dong, Weiqing He, Shuyi Si, Bin Hong, Yinghong Li, Danqing Song, Zonggen Peng, Yongsheng Che, Jian-Dong Jiang. Carrimycin inhibits coronavirus replication by decreasing the efficiency of programmed –1 ribosomal frameshifting through directly binding to the RNA pseudoknot of viral frameshift-stimulatory element. Acta Pharmaceutica Sinica B 2024, 14
(6)
, 2567-2580. https://doi.org/10.1016/j.apsb.2024.02.023
- Ajay Vikram Singh, Preeti Bhardwaj, Aditya Kumar Upadhyay, Anselmo Pagani, Jyoti Upadhyay, Jolly Bhadra, Veronica Tisato, Manali Thakur, Donato Gemmati, Rudresh Mishra, Paolo Zamboni. Navigating regulatory challenges in molecularly tailored nanomedicine. Exploration of BioMat-X 2024, 1
(2)
, 124-134. https://doi.org/10.37349/ebmx.2024.00009
- Nikolay T. Tzvetkov, Martina I. Peeva, Maya G. Georgieva, Vera Deneva, Aneliya A. Balacheva, Ivan P. Bogdanov, Maria Ponticelli, Luigi Milella, Kiril Kirilov, Maima Matin, Hans-Georg Stammler, Atanas G. Atanasov, Liudmil Antonov. Favipiravir vs. Deferiprone: Tautomeric, photophysical, in vitro biological studies, and binding interactions with SARS-Cov-2-MPro/ACE2. Current Research in Biotechnology 2024, 7 , 100176. https://doi.org/10.1016/j.crbiot.2024.100176
- Wenhui Lu, Xiaoying Li, Wenyi You, Rui Gong. Rhabdomyolysis in a patient with end-stage renal disease and SARS-CoV-2 infection: A case report. Medicine 2023, 102
(48)
, e36360. https://doi.org/10.1097/MD.0000000000036360
- Mufarreh Asmari, Lenka Michalcová, Adel Ehab Ibrahim, Zdeněk Glatz, Hermann Wätzig, Sami El Deeb. Studying molecular interactions via capillary electrophoresis and microscale thermophoresis: A review. ELECTROPHORESIS 2023, 44
(13-14)
, 1114-1142. https://doi.org/10.1002/elps.202200275
- Alan J. Waring, Grace C.-L. Jung, Shantanu K. Sharma, Frans J. Walther. Lung Surfactant Protein B Peptide Mimics Interact with the Human ACE2 Receptor. International Journal of Molecular Sciences 2023, 24
(13)
, 10837. https://doi.org/10.3390/ijms241310837
- Jing-Fang Yang, Fan Wang, Meng-Yao Wang, Di Wang, Zhong-Shi Zhou, Ge-Fei Hao, Qing X. Li, Guang-Fu Yang. CIPDB: A biological structure databank for studying cation and π interactions. Drug Discovery Today 2023, 28
(5)
, 103546. https://doi.org/10.1016/j.drudis.2023.103546
- Alessandra Monti, Luigi Vitagliano, Andrea Caporale, Menotti Ruvo, Nunzianna Doti. Targeting Protein–Protein Interfaces with Peptides: The Contribution of Chemical Combinatorial Peptide Library Approaches. International Journal of Molecular Sciences 2023, 24
(9)
, 7842. https://doi.org/10.3390/ijms24097842
- Kelly Bugatti, Andrea Sartori, Lucia Battistini, Crescenzo Coppa, Emiel Vanhulle, Sam Noppen, Becky Provinciael, Lieve Naesens, Annelies Stevaert, Alessandro Contini, Kurt Vermeire, Franca Zanardi. Novel Polymyxin-Inspired Peptidomimetics Targeting the SARS-CoV-2 Spike:hACE2 Interface. International Journal of Molecular Sciences 2023, 24
(10)
, 8765. https://doi.org/10.3390/ijms24108765
- Nikolay Tzvetkov Tzvetkov, Martina Ivova Peeta, Maya Georgieva Georgieva, Vera Deneva, Aneliya Anatolieva Balacheva, Ivan Petrov Bogdanov, Maria Ponticelli, Luigi Milella, Kiril Kirilov, Maima Matin, Hans-Georg Stammler, Atanas Georgiev Atanasov, Liudmil Antonov. Favipiravir vs. Deferiprone: Tautomeric, Photophysical, in vitro Biological Evaluation and Binding Interactions with SARS-CoV-2MPro/ACE2. 2023https://doi.org/10.2139/ssrn.4614286
- MOHAMAD ZULKEFLEE SABRI, JOANNA BOJARSKA, FAI-CHU WONG, TSUN-THAI CHAI. Molecular dynamics-driven exploration of peptides targeting SARS-CoV-2, with special attention on ACE2, S protein, Mpro, and PLpro: A review. BIOCELL 2023, 47
(8)
, 1727-1742. https://doi.org/10.32604/biocell.2023.029272
- Ran Zhang, Kai Gao, Afsaneh Sadremomtaz, Angel J. Ruiz-Moreno, Alessandra Monti, Zayana M. Al-Dahmani, Ben Gyau, Nunzianna Doti, Matthew R. Groves. Identification of Hotspots in Synthetic Peptide Inhibitors of the FOXO4:p53 Interaction. 2022https://doi.org/10.21203/rs.3.rs-2361374/v1
- M. V. Sidorova, R. S. Bibilashvili, D. V. Avdeev, U. S. Kozhokar, M. E. Palkeeva, M. V. Ovchinnikov, A. S. Molokoedov, D. A. Shirokov, A. V. Semyonova, V. I. Uvarova, P. O. Kulyaev, E. V. Khvatov, A. A. Ignatova, A. V. Feofanov, D. I. Osolodkin, Yu. B. Porozov, L. I. Kozlovskaya, A. A. Ishmukhametov, Ye. V. Parfyonova, A. M. Egorov. Properties and Activity of Peptide Derivatives of ACE2 Cellular Receptor and Their Interaction with SARS-CoV-2 S Protein Receptor-Binding Domain. Doklady Biochemistry and Biophysics 2022, 507
(1)
, 237-241. https://doi.org/10.1134/S1607672922060126
- 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
- Carla Zannella, Annalisa Chianese, Giuseppe Greco, Biagio Santella, Giuseppe Squillaci, Alessandra Monti, Nunzianna Doti, Giuseppina Sanna, Aldo Manzin, Alessandra Morana, Anna De Filippis, Gianni D’Angelo, Francesco Palmieri, Gianluigi Franci, Massimiliano Galdiero. Design of Three Residues Peptides against SARS-CoV-2 Infection. Viruses 2022, 14
(10)
, 2103. https://doi.org/10.3390/v14102103
- Salman Ali Khan, Alamgir Khan, Komal Zia, Ihab Shawish, Assem Barakat, Zaheer Ul-Haq. Cheminformatics-Based Discovery of Potential Chemical Probe Inhibitors of Omicron Spike Protein. International Journal of Molecular Sciences 2022, 23
(18)
, 10315. https://doi.org/10.3390/ijms231810315
- Lucas Weißenborn, Elie Richel, Helena Hüseman, Julia Welzer, Silvan Beck, Simon Schäfer, Heinrich Sticht, Klaus Überla, Jutta Eichler. Smaller, Stronger, More Stable: Peptide Variants of a SARS-CoV-2 Neutralizing Miniprotein. International Journal of Molecular Sciences 2022, 23
(11)
, 6309. https://doi.org/10.3390/ijms23116309
- Mark D. Nolan, Andrea Mezzetta, Lorenzo Guazzelli, Eoin M. Scanlan. Radical-mediated thiol–ene ‘click’ reactions in deep eutectic solvents for bioconjugation. Green Chemistry 2022, 24
(4)
, 1456-1462. https://doi.org/10.1039/D1GC03714E
- Akshay Chenna, Wajihul H Khan, Rozaleen Dash, Anurag S Rathore, Gaurav Goel. Template-based design of peptides to inhibit SARS-CoV-2 RNA-dependent RNA polymerase complexation. 2022https://doi.org/10.1101/2022.01.24.477502
- Ying-Chiang J. Lee, Jaden D. Shirkey, Jongbeom Park, Karishma Bisht, Alexis J. Cowan. An Overview of Antiviral Peptides and Rational Biodesign Considerations. BioDesign Research 2022, 2022 , 9898241. https://doi.org/10.34133/2022/9898241
- Narcis Fernandez-Fuentes, Ruben Molina, Baldo Oliva. A Collection of Designed Peptides to Target SARS-CoV-2 Spike RBD—ACE2 Interaction. International Journal of Molecular Sciences 2021, 22
(21)
, 11627. https://doi.org/10.3390/ijms222111627
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
Abstract
Figure 1
Figure 1. Interaction of ACE2 with S-RBD. (a) Surface representation of the complex between the receptor binding (S-RBD) domain of SARS-CoV-2 Spike protein (yellow) and the human ACE2 receptor (pink) (PDB ID: 6M0J). The portion of the ACE2 domain including main interacting residues of helices α1 (I21, Q24, T27, F28, D30, K31, H34, E35, E37, D38, Y41, Q42), α2 (L79, M82, Y83), and α3 (N330, K419, D430, E431) and β sheets β3 and β4 (K353, G354, D355, and R357) are drawn in green. (b) A closer view displays the interacting residues at the interface site. Figure created by PyMol (Molecular Graphics System, ver. 1.2r3pre, Schrödinger, LLC).
Figure 2
Figure 2. Stick representation of residues involved in the interprotomer interaction of S-RBD. (a) Side view of the surface representation of the interactions within ACE2 and S-RRBD (PDB ID: 6M0J). (b) Residues involved in the subunit interaction are shown in green (cartoon transparency is set to 40%). Four contact regions are located in the α1, α2and α3 helices and in β3 and β4 of ACE2 and S-RBD. Figure created by PyMol (Molecular Graphics System, ver. 1.2r3pre, Schrödinger, LLC).
Figure 3
Figure 3. (a) Heat maps representing the pairwise backbone RMSD matrix of SARS-CoV-2 S-RBD protein calculated for the backbone along 50 ns of MD simulation from systems including peptides with stable binding to S-RBD. The unliganded protein (apo-RBD) is included for comparison. The simulation corresponding to apo-RBD displays a higher RMSD in comparison with the matrices originated for peptides 1, 2, 5, and 6. Indicating a less flexible conformation of S-RBD. (b) The unliganded protein (ACE2:S-RBD) is included for comparison
Figure 4
Figure 4. α-Carbon RMSF analysis for the peptide-S-RBD systems. (a) α-Carbon RMSF profiles of all studied peptides, apo-S-RBD, and ACE2 are presented for comparison. (b) RMSF structural representation of apo-S-RBD and ACE2:S-RBD. (c) structural representation of peptide:S-RBD complexes including the binding of the peptides across 10 representative snapshots. Normalized scale for peptides 1–5; peptide 6 is presented with its own scale.
Figure 5
Figure 5. (a) Binding analysis for the interaction between S-RBD and ACE2. (a) Luciferase-based assay. 293T cells were transfected with the ACE2 or S-RBD expression constructs. 48 h post-transfection, and luciferase assays were performed on 20 μg total protein from cell lysates using FMZ as a substrate (n = 3, mean ± SD; one-way ANOVA, ***p < 0.005 relative to smBiT-ACE2 alone, Dunnett’s correction for multiple comparisons). (b) MST analysis of peptide 1–6 binding to recombinant S-RBD. The concentration of S-RBD is kept constant at 50 nM, while the ligand concentration varies from 12.5 μM to 0.19 nM. Serial titrations result in measurable changes in the fluorescence signal within a temperature gradient that can be used to calculate the dissociation constant (Kd). The curve is shown as ΔFnorm (change of Fnorm with respect to the zero-ligand concentration) against S-RBD concentration on a log scale.
References
This article references 77 other publications.
- 1Liu, C. H.; Lu, C. H.; Wong, S. H.; Lin, L. T. Update on antiviral strategies against COVID-19: unmet needs and prospects. Front. Immunol. 2021, 11, 616595, DOI: 10.3389/fimmu.2020.616595There is no corresponding record for this reference.
- 2Galindez, G.; Matschinske, J.; Rose, T. D.; Sadegh, S.; Salgado-Albarrán, M.; Späth, J.; Baumbach, J.; Pauling, J. K. Lessons from the COVID-19 pandemic for advancing computational drug repurposing strategies. Nat. Comput. Sci. 2021, 1, 33– 41, DOI: 10.1038/s43588-020-00007-6There is no corresponding record for this reference.
- 3Hufsky, F.; Lamkiewicz, K.; Almeida, A.; Aouacheria, A.; Arighi, C. Computational strategies to combat COVID-19: useful tools to accelerate SARS-CoV-2 and coronavirus research. Briefings Bioinf. 2021, 22, 642– 663, DOI: 10.1093/bib/bbaa2323https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsFWht73E&md5=b79ea64cf7ce9c512e9481ea30416bc8Computational strategies to combat COVID-19: useful tools to accelerate SARS-CoV-2 and coronavirus researchHufsky, Franziska; Lamkiewicz, Kevin; Almeida, Alexandre; Aouacheria, Abdel; Arighi, Cecilia; Bateman, Alex; Baumbach, Jan; Beerenwinkel, Niko; Brandt, Christian; Cacciabue, Marco; Chuguransky, Sara; Drechsel, Oliver; Finn, Robert D.; Fritz, Adrian; Fuchs, Stephan; Hattab, Georges; Hauschild, Anne-Christin; Heider, Dominik; Hoffmann, Marie; Holzer, Martin; Hoops, Stefan; Kaderali, Lars; Kalvari, Ioanna; von Kleist, Max; Kmiecinski, Reno; Kuhnert, Denise; Lasso, Gorka; Libin, Pieter; List, Markus; Lochel, Hannah F.; Martin, Maria J.; Martin, Roman; Matschinske, Julian; McHardy, Alice C.; Mendes, Pedro; Mistry, Jaina; Navratil, Vincent; Nawrocki, Eric P.; O'Toole, Aine Niamh; Ontiveros-Palacios, Nancy; Petrov, Anton I.; Rangel-Pineros, Guillermo; Redaschi, Nicole; Reimering, Susanne; Reinert, Knut; Reyes, Alejandro; Richardson, Lorna; Robertson, David L.; Sadegh, Sepideh; Singer, Joshua B.; Theys, Kristof; Upton, Chris; Welzel, Marius; Williams, Lowri; Marz, ManjaBriefings in Bioinformatics (2021), 22 (2), 642-663CODEN: BBIMFX; ISSN:1477-4054. (Oxford University Press)A review. SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) is a novel virus of the family Coronaviridae. The virus causes the infectious disease COVID-19. The biol. of coronaviruses has been studied for many years. However, bioinformatics tools designed explicitly for SARS-CoV-2 have only recently been developed as a rapid reaction to the need for fast detection, understanding and treatment of COVID-19. To control the ongoing COVID-19 pandemic, it is of utmost importance to get insight into the evolution and pathogenesis of the virus. In this review, we cover bioinformatics workflows and tools for the routine detection of SARS-CoV-2 infection, the reliable anal. of sequencing data, the tracking of the COVID-19 pandemic and evaluation of containment measures, the study of coronavirus evolution, the discovery of potential drug targets and development of therapeutic strategies. For each tool, we briefly describe its use case and how it advances research specifically for SARS-CoV-2. All tools are free to use and available online, either through web applications or public code repositories.
- 4V’kovski, P.; Kratzel, A.; Steiner, S.; Stalder, H.; Thiel, V. Coronavirus biology and replication: implications for SARS-CoV-2. Nat. Rev. Microbiol. 2021, 19, 155– 170, DOI: 10.1038/s41579-020-00468-64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1GjsLjF&md5=14ddf54f3e569fe39eb4f3295cf603e6Coronavirus biology and replication: implications for SARS-CoV-2V'kovski, Philip; Kratzel, Annika; Steiner, Silvio; Stalder, Hanspeter; Thiel, VolkerNature Reviews Microbiology (2021), 19 (3), 155-170CODEN: NRMACK; ISSN:1740-1526. (Nature Research)A review. Abstr.: The SARS-CoV-2 pandemic and its unprecedented global societal and economic disruptive impact has marked the third zoonotic introduction of a highly pathogenic coronavirus into the human population. Although the previous coronavirus SARS-CoV and MERS-CoV epidemics raised awareness of the need for clin. available therapeutic or preventive interventions, to date, no treatments with proven efficacy are available. The development of effective intervention strategies relies on the knowledge of mol. and cellular mechanisms of coronavirus infections, which highlights the significance of studying virus-host interactions at the mol. level to identify targets for antiviral intervention and to elucidate crit. viral and host determinants that are decisive for the development of severe disease. In this Review, we summarize the first discoveries that shape our current understanding of SARS-CoV-2 infection throughout the intracellular viral life cycle and relate that to our knowledge of coronavirus biol. The elucidation of similarities and differences between SARS-CoV-2 and other coronaviruses will support future preparedness and strategies to combat coronavirus infections.
- 5Zhang, 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 α-ketoamide inhibitors. Science 2020, 368, 409– 412, DOI: 10.1126/science.abb34055https://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.
- 6Jin, 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 Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289– 293, DOI: 10.1038/s41586-020-2223-y6https://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.
- 7Hussain, S.; Xie, Y. J.; Li, D.; Malik, S. I.; Hou, J. C.; Leung, E. L.; Fan, X. X. Current strategies against COVID-19. Chin. Med. 2020, 15, 70, DOI: 10.1186/s13020-020-00353-77https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlOhurzJ&md5=a8d06ec5240ed4d5224dfb3be7c76635Current strategies against COVID-19Hussain, Shahid; Xie, Ya-Jia; Li, Dan; Malik, Shaukat Iqbal; Hou, Jin-cai; Leung, Elaine Lai-Han; Fan, Xing-XingChinese Medicine (London, United Kingdom) (2020), 15 (1), 70CODEN: CMHEBK; ISSN:1749-8546. (BioMed Central Ltd.)A review. Abstr.: Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) recently was declared a pandemic by world health organization (WHO) Due to sudden outbreaks, currently, no completely effective vaccine or drug is clin. approved. Several therapeutic strategies can be envisaged to prevent further mortality and morbidity. Based on the past contribution of traditional Chinese medicines (TCM) and immune-based therapies as a treatment option in crucial pathogen outbreaks, we aimed to summarize potential therapeutic strategies that could be helpful to stop further spread of SARS-CoV-2 by effecting its structural components or modulation of immune responses. A review. Several TCM with or without modification could be effective against the structural protein, enzymes, and nucleic acid should be tested from available libraries or to identify their immune-stimulatory activities to enhance several antiviral biol. agents for effective elimination of SARS-CoV-2 from the host. TCM is not only effective in the direct inhibition of virus attachment and internalization in a cell but can also prevent their replication and can also help to boost up host immune response. Immune-modulatory effects of TCMs may lead to new medications and can guide us for the scientific validity of drug development. Besides, we also summarized the effective therapies in clin. for controlling inflammation. This review will be not only helpful for the current situation of COVID-19, but can also play a major role in such epidemics in the future.
- 8Choudhary, S.; Malik, Y. S.; Tomar, S. Identification of SARS-CoV-2 cell entry inhibitors by drug repurposing using in silico structure-based virtual screening approach. Front. Immunol. 2020, 11, 1664, DOI: 10.3389/fimmu.2020.016648https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlSis7vM&md5=dc9eff66bcebb7f5a900170511eef385Identification of SARS-CoV-2 cell entry inhibitors by drug repurposing using in silico structure-based virtual screening approachChoudhary, Shweta; Malik, Yashpal S.; Tomar, ShaillyFrontiers in Immunology (2020), 11 (), 1664CODEN: FIRMCW; ISSN:1664-3224. (Frontiers Media S.A.)The rapidly spreading, highly contagious and pathogenic SARS-coronavirus 2 (SARS-CoV-2) assocd. Coronavirus Disease 2019 (COVID-19) has been declared as a pandemic by the World Health Organization (WHO). The novel 2019 SARS-CoV-2 enters the host cell by binding of the viral surface spike glycoprotein (S-protein) to cellular angiotensin converting enzyme 2 (ACE2) receptor. The virus specific mol. interaction with the host cell represents a promising therapeutic target for identifying SARS-CoV-2 antiviral drugs. The repurposing of drugs can provide a rapid and potential cure toward exponentially expanding COVID-19. Thereto, high throughput virtual screening approach was used to investigate FDA approved LOPAC library drugs against both the receptor binding domain of spike protein (S-RBD) and ACE2 host cell receptor. Primary screening identified a few promising mols. for both the targets, which were further analyzed in details by their binding energy, binding modes through mol. docking, dynamics and simulations. Evidently, GR 127935 hydrochloride hydrate, GNF-5, RS504393, TNP, and eptifibatide acetate were found binding to virus binding motifs of ACE2 receptor. Addnl., KT203, BMS195614, KT185, RS504393, and GSK1838705A were identified to bind at the receptor binding site on the viral S-protein. These identified mols. may effectively assist in controlling the rapid spread of SARS-CoV-2 by not only potentially inhibiting the virus at entry step but are also hypothesized to act as anti-inflammatory agents, which could impart relief in lung inflammation. Timely identification and detn. of an effective drug to combat and tranquilize the COVID-19 global crisis is the utmost need of hour. Further, prompt in vivo testing to validate the anti-SARS-CoV-2 inhibition efficiency by these mols. could save lives is justified.
- 9Shyr, Z. A.; Gorshkov, K.; Chen, C. Z.; Zheng, W. Drug discovery strategies for SARS-CoV-2. J. Pharmacol. Exp. Ther. 2020, 375, 127– 138, DOI: 10.1124/jpet.120.0001239https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1Kis7jE&md5=c0bfab352eefb70fe2b137a8b7d3899fDrug discovery strategies for SARS-CoV-2Shyr, Zeenat A.; Gorshkov, Kirill; Chen, Catherine Z.; Zheng, WeiJournal of Pharmacology and Experimental Therapeutics (2020), 375 (1), 127-138CODEN: JPETAB; ISSN:1521-0103. (American Society for Pharmacology and Experimental Therapeutics)A review. Coronavirus disease 2019 (COVID-19) is a novel disease caused by the severe acute respiratory syndrome coronavirus (SARS-CoV)-2 virus that was first detected in Dec. of 2019 in Wuhan, China, and has rapidly spread worldwide. The search for a suitable vaccine as well as effective therapeutics for the treatment of COVID-19 is underway. Drug repurposing screens provide a useful and effective soln. for identifying potential therapeutics against SARS-CoV-2. For example, the exptl. drug remdesivir, originally developed for Ebola virus infections, has been approved by the US Food and Drug Administration as an emergency use treatment of COVID-19. However, the efficacy and toxicity of this drug need further improvements. In this review, we discuss recent findings on the pathol. of coronaviruses and the drug targets for the treatment of COVID-19. Both SARS-CoV-2-specific inhibitors and broad-spectrum anticoronavirus drugs against SARS-CoV, Middle East respiratory syndrome coronavirus, and SARS-CoV-2 will be valuable addns. to the anti-SARS-CoV-2 armament. A multitarget treatment approach with synergistic drug combinations contg. different mechanisms of action may be a practical therapeutic strategy for the treatment of severe COVID-19.
- 10Gil, C.; Ginex, T.; Maestro, I.; Nozal, V.; Barrado-Gil, L.; Cuesta-Geijo, M.Á.; Urquiza, J.; Ramírez, D.; Alonso, C.; Campillo, N. E.; Martinez, A. COVID-19: drug targets and potential treatments. J. Med. Chem. 2020, 63, 12359– 12386, DOI: 10.1021/acs.jmedchem.0c0060610https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFWmsL3I&md5=6a0dc17c5f601f09a92da933f81bbbeaCOVID-19: Drug Targets and Potential TreatmentsGil, Carmen; Ginex, Tiziana; Maestro, Ines; Nozal, Vanesa; Barrado-Gil, Lucia; Cuesta-Geijo, Miguel Angel; Urquiza, Jesus; Ramirez, David; Alonso, Covadonga; Campillo, Nuria E.; Martinez, AnaJournal of Medicinal Chemistry (2020), 63 (21), 12359-12386CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. Currently, the authors are immersed in a pandemic caused by the emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which severely threatens public health worldwide. Until now, no drug or vaccine has been approved to treat the severe disease caused by this coronavirus, COVID-19. The authors will focus on the main virus-based and host-based targets that can guide medicinal chem. efforts to discover new drugs for this devastating disease. In principle, all CoVs enzymes and proteins involved in viral replication and the control of host cellular machineries are potentially druggable targets in the search for therapeutic options for SARS-CoV-2. This perspective provides an overview of the main targets from a structural point of view, together with reported therapeutic compds. with activity against SARS-CoV-2 and/or other CoVs. Also, the role of innate immune response to coronavirus infection and the related therapeutic options will be presented.
- 11Catanzaro, M.; Fagiani, F.; Racchi, M.; Corsini, E.; Govoni, S.; Lanni, C. Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transduct Target Ther. 2020, 5, 84, DOI: 10.1038/s41392-020-0191-111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38rivVOmuw%253D%253D&md5=3fb8e4f85321d6225c5c4922661eab10Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2Catanzaro Michele; Fagiani Francesca; Racchi Marco; Govoni Stefano; Lanni Cristina; Fagiani Francesca; Corsini EmanuelaSignal transduction and targeted therapy (2020), 5 (1), 84 ISSN:.To date, no vaccines or effective drugs have been approved to prevent or treat COVID-19 and the current standard care relies on supportive treatments. Therefore, based on the fast and global spread of the virus, urgent investigations are warranted in order to develop preventive and therapeutic drugs. In this regard, treatments addressing the immunopathology of SARS-CoV-2 infection have become a major focus. Notably, while a rapid and well-coordinated immune response represents the first line of defense against viral infection, excessive inflammatory innate response and impaired adaptive host immune defense may lead to tissue damage both at the site of virus entry and at systemic level. Several studies highlight relevant changes occurring both in innate and adaptive immune system in COVID-19 patients. In particular, the massive cytokine and chemokine release, the so-called "cytokine storm", clearly reflects a widespread uncontrolled dysregulation of the host immune defense. Although the prospective of counteracting cytokine storm is compelling, a major limitation relies on the limited understanding of the immune signaling pathways triggered by SARS-CoV-2 infection. The identification of signaling pathways altered during viral infections may help to unravel the most relevant molecular cascades implicated in biological processes mediating viral infections and to unveil key molecular players that may be targeted. Thus, given the key role of the immune system in COVID-19, a deeper understanding of the mechanism behind the immune dysregulation might give us clues for the clinical management of the severe cases and for preventing the transition from mild to severe stages.
- 12Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X.; Zheng, M.; Chen, L.; Li, H. Analysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methods. Acta Pharm. Sin. B 2020, 10, 766– 788, DOI: 10.1016/j.apsb.2020.02.00812https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXnslWnug%253D%253D&md5=e644ad7f22b6780443864db76255c8fcAnalysis of therapeutic targets for SARS-CoV-2 and discovery of potential drugs by computational methodsWu, Canrong; Liu, Yang; Yang, Yueying; Zhang, Peng; Zhong, Wu; Wang, Yali; Wang, Qiqi; Xu, Yang; Li, Mingxue; Li, Xingzhou; Zheng, Mengzhu; Chen, Lixia; Li, HuaActa Pharmaceutica Sinica B (2020), 10 (5), 766-788CODEN: APSBCW; ISSN:2211-3835. (Elsevier B.V.)SARS-CoV-2 has caused tens of thousands of infections and more than one thousand deaths. There are currently no registered therapies for treating coronavirus infections. Because of time consuming process of new drug development, drug repositioning may be the only soln. to the epidemic of sudden infectious diseases. We systematically analyzed all the proteins encoded by SARS-CoV-2 genes, compared them with proteins from other coronaviruses, predicted their structures, and built 19 structures that could be done by homol. modeling. By performing target-based virtual ligand screening, a total of 21 targets (including two human targets) were screened against compd. libraries including ZINC drug database and our own database of natural products. Structure and screening results of important targets such as 3-chymotrypsin-like protease (3CLpro), Spike, RNA-dependent RNA polymerase (RdRp), and papain like protease (PLpro) were discussed in detail. In addn., a database of 78 commonly used anti-viral drugs including those currently on the market and undergoing clin. trials for SARS-CoV-2 was constructed. Possible targets of these compds. and potential drugs acting on a certain target were predicted. This study will provide new lead compds. and targets for further in vitro and in vivo studies of SARS-CoV-2, new insights for those drugs currently ongoing clin. studies, and also possible new strategies for drug repositioning to treat SARS-CoV-2 infections.
- 13VanPatten, S.; He, M.; Altiti, A.; F Cheng, K.; Ghanem, M. H.; Al-Abed, Y. Evidence supporting the use of peptides and peptidomimetics as potential SARS-CoV-2 (COVID-19) therapeutics. Future Med. Chem. 2020, 12, 1647– 1656, DOI: 10.4155/fmc-2020-018013https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvV2nsLzM&md5=de5929a158d71c2f4f8039ebff146307Evidence supporting the use of peptides and peptidomimetics as potential SARS-CoV-2 (COVID-19) therapeuticsVanPatten, Sonya; He, Mingzhu; Altiti, Ahmad; F Cheng, Kai; Ghanem, Mustafa H.; Al-Abed, YousefFuture Medicinal Chemistry (2020), 12 (18), 1647-1656CODEN: FMCUA7; ISSN:1756-8919. (Newlands Press Ltd.)A review. During a disease outbreak/pandemic situation such as COVID-19, researchers are in a prime position to identify and develop peptide-based therapies, which could be more rapidly and cost-effectively advanced into a clin. setting. One drawback of natural peptide drugs, however, is their proteolytic instability; peptidomimetics can help to overcome this caveat. In this review, we summarize peptide and peptide-based therapeutics that target one main entry pathway of SARS-CoV-2, which involves the host ACE2 receptor and viral spike (S) protein interaction. Furthermore, we discuss the advantages of peptidomimetics and other potential targets that have been studied using peptide-based therapeutics for COVID-19.
- 14Smith, M. C.; Gestwicki, J. E. Features of protein-protein interactions that translate into potent inhibitors: topology, surface area and affinity. Expert Rev. Mol. Med. 2012, 14, 14– 16, DOI: 10.1017/erm.2012.10There is no corresponding record for this reference.
- 15Josephson, K.; Ricardo, A.; Szostak, J. W. mRNA display: from basic principles to macrocycle drug discovery. Drug Discovery Today 2014, 19, 388– 99, DOI: 10.1016/j.drudis.2013.10.01115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsleltrnI&md5=6676fef2ca8f0a81cc0e993ee4ec7424mRNA display: from basic principles to macrocycle drug discoveryJosephson, Kristopher; Ricardo, Alonso; Szostak, Jack W.Drug Discovery Today (2014), 19 (4), 388-399CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. We describe a new discovery technol. that uses mRNA-display to rapidly synthesize and screen macrocyclic peptide libraries to explore a valuable region of chem. space typified by natural products. This technol. allows high-affinity peptidic macrocycles contg. modified backbones and unnatural side chains to be readily selected based on target binding. Success stories covering the first examples of these libraries suggest that they could be used for the discovery of intracellular protein-protein interaction inhibitors, highly selective enzyme inhibitors or synthetic replacements for monoclonal antibodies. The review concludes with a look to the future regarding how this technol. might be improved with respect to library design for cell permeability and bioavailability.
- 16WHO. Draft landscape of COVID-19 candidate vaccines. https://www.who.int/news-room/q-a-detail/coronavirus-disease-(covid-19)-vaccines (accessed January 2021).There is no corresponding record for this reference.
- 17Voysey, M.; Clemens; Madhi, S.; Weckx, L. Y.; Folegatti, P. M. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet 2021, 397, 99– 111, DOI: 10.1016/S0140-6736(20)32661-117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFCgt77I&md5=826fc2243914f0e5bcdf0931520c2b42Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomized controlled trials in Brazil, South Africa, and the UKVoysey, Merryn; Clemens, Sue Ann Costa; Madhi, Shabir A.; Weckx, Lily Y.; Folegatti, Pedro M.; Aley, Parvinder K.; Angus, Brian; Baillie, Vicky L.; Barnabas, Shaun L.; Bhorat, Qasim E.; Bibi, Sagida; Briner, Carmen; Cicconi, Paola; Collins, Andrea M.; Colin-Jones, Rachel; Cutland, Clare L.; Darton, Thomas C.; Dheda, Keertan; Duncan, Christopher J. A.; Emary, Katherine R. W.; Ewer, Katie J.; Fairlie, Lee; Faust, Saul N.; Feng, Shuo; Ferreira, Daniela M.; Finn, Adam; Goodman, Anna L.; Green, Catherine M.; Green, Christopher A.; Heath, Paul T.; Hill, Catherine; Hill, Helen; Hirsch, Ian; Hodgson, Susanne H. C.; Izu, Alane; Jackson, Susan; Jenkin, Daniel; Joe, Carina C. D.; Kerridge, Simon; Koen, Anthonet; Kwatra, Gaurav; Lazarus, Rajeka; Lawrie, Alison M.; Lelliott, Alice; Libri, Vincenzo; Lillie, Patrick J.; Mallory, Raburn; Mendes, Ana V. A.; Milan, Eveline P.; Minassian, Angela M.; McGregor, Alastair; Morrison, Hazel; Mujadidi, Yama F.; Nana, Anusha; O'Reilly, Peter J.; Padayachee, Sherman D.; Pittella, Ana; Plested, Emma; Pollock, Katrina M.; Ramasamy, Maheshi N.; Rhead, Sarah; Schwarzbold, Alexandre V.; Singh, Nisha; Smith, Andrew; Song, Rinn; Snape, Matthew D.; Sprinz, Eduardo; Sutherland, Rebecca K.; Tarrant, Richard; Thomson, Emma C.; Torok, M. Estee; Toshner, Mark; Turner, David P. J.; Vekemans, Johan; Villafana, Tonya L.; Watson, Marion E. E.; Williams, Christopher J.; Douglas, Alexander D.; Hill, Adrian V. S.; Lambe, Teresa; Gilbert, Sarah C.; Pollard, Andrew J.Lancet (2021), 397 (10269), 99-111CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A safe and efficacious vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), if deployed with high coverage, could contribute to the control of the COVID-19 pandemic. We evaluated the safety and efficacy of the ChAdOx1 nCoV-19 vaccine in a pooled interim anal. of four trials. This anal. includes data from four ongoing blinded, randomized, controlled trials done across the UK, Brazil, and South Africa. Participants aged 18 years and older were randomly assigned (1:1) to ChAdOx1 nCoV-19 vaccine or control (meningococcal group A, C, W, and Y conjugate vaccine or saline). Participants in the ChAdOx1 nCoV-19 group received two doses contg. 5 x 1010 viral particles (std. dose; SD/SD cohort); a subset in the UK trial received a half dose as their first dose (low dose) and a std. dose as their second dose (LD/SD cohort). The primary efficacy anal. included symptomatic COVID-19 in seroneg. participants with a nucleic acid amplification test-pos. swab more than 14 days after a second dose of vaccine. Participants were analyzed according to treatment received, with data cutoff on Nov 4, 2020. Vaccine efficacy was calcd. as 1 - relative risk derived from a robust Poisson regression model adjusted for age. Studies are registered at ISRCTN89951424 and ClinicalTrials.gov, NCT04324606, NCT04400838, and NCT04444674. Between Apr. 23 and Nov 4, 2020, 23,848 participants were enrolled and 11,636 participants (7548 in the UK, 4088 in Brazil) were included in the interim primary efficacy anal. In participants who received two std. doses, vaccine efficacy was 62.1% (95% CI 41.0-75.7; 27 [0.6%] of 4440 in the ChAdOx1 nCoV-19 group vs. 71 [1.6%] of 4455 in the control group) and in participants who received a low dose followed by a std. dose, efficacy was 90.0% (67.4-97.0; three [0.2%] of 1367 vs 30 [2.2%] of 1374; pinteraction=0.010). Overall vaccine efficacy across both groups was 70.4% (95.8% CI 54.8-80.6; 30 [0.5%] of 5807 vs 101 [1.7%] of 5829). From 21 days after the first dose, there were ten cases hospitalized for COVID-19, all in the control arm; two were classified as severe COVID-19, including one death. There were 74,341 person-months of safety follow-up (median 3.4 mo, IQR 1.3-4.8): 175 severe adverse events occurred in 168 participants, 84 events in the ChAdOx1 nCoV-19 group and 91 in the control group. Three events were classified as possibly related to a vaccine: one in the ChAdOx1 nCoV-19 group, one in the control group, and one in a participant who remains masked to group allocation. ChAdOx1 nCoV-19 has an acceptable safety profile and has been found to be efficacious against symptomatic COVID-19 in this interim anal. of ongoing clin. trials.
- 18Polack, F. P.; Thomas, S. J.; Kitchin, N. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N. Engl. J. Med. 2020, 383, 2603– 2615, DOI: 10.1056/NEJMoa203457718https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXotFSjuw%253D%253D&md5=08cb7204334ed37c7949dfcdfe5af75cSafety and efficacy of the BNT162b2 mRNA Covid-19 vaccinePolack, Fernando P.; Thomas, Stephen J.; Kitchin, Nicholas; Absalon, Judith; Gurtman, Alejandra; Lockhart, Stephen; Perez, John L.; Marc, Gonzalo Perez; Moreira, Edson D.; Zerbini, Cristiano; Bailey, Ruth; Swanson, Kena A.; Roychoudhury, Satrajit; Koury, Kenneth; Li, Ping; Kalina, Warren V.; Cooper, David; Frenck, Robert W., Jr.; Hammitt, Laura L.; Tureci, Ozlem; Nell, Haylene; Schaefer, Axel; Unal, Serhat; Tresnan, Dina B.; Mather, Susan; Dormitzer, Philip R.; Sahin, Ugur; Jansen, Kathrin U.; Gruber, William C.New England Journal of Medicine (2020), 383 (27), 2603-2615CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)A review. Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a worldwide pandemic. Safe and effective vaccines are needed urgently. methods In an ongoing multinational, placebo-controlled, observer-blinded, pivotal efficacy trial, we randomly assigned persons 16 years of age or older in a 1:1 ratio to receive two doses, 21 days apart, of either placebo or the BNT162b2 vaccine candidate (30μg per dose). BNT162b2 is a lipid nanoparticle-formulated, nucleoside-modified RNA vaccine that encodes a prefusion stabilized, membrane-anchored SARS-CoV-2 full-length spike protein. The primary end points were efficacy of the vaccine against lab.-confirmed Covid-19 and safety. results A total of 43,548 participants underwent randomization, of whom 43,448 received injections: 21,720 with BNT162b2 and 21,728 with placebo. There were 8 cases of Covid-19 with onset at least 7 days after the second dose among participants assigned to receive BNT162b2 and 162 cases among those assigned to placebo; BNT162b2 was 95% effective in preventing Covid-19 (95% credible interval, 90.3 to 97.6). Similar vaccine efficacy (generally 90 to 100%) was obsd. across subgroups defined by age, sex, race, ethnicity, baseline body-mass index, and the presence of coexisting conditions. Among 10 cases of severe Covid-19 with onset after the first dose, 9 occurred in placebo recipients and 1 in a BNT162b2 recipient. The safety profile of BNT162b2 was characterized by short-term, mild-to-moderate pain at the injection site, fatigue, and headache. The incidence of serious adverse events was low and was similar in the vaccine and placebo groups. conclusions A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older. Safety over a median of 2 mo was similar to that of other viral vaccines.
- 19Baden, L. R.; El Sahly, H. M.; Essink, B. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 2021, 384, 403– 416, DOI: 10.1056/NEJMoa203538919https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFSmsrc%253D&md5=9b0c79f3e3fd2ea452a3fbf57281720fEfficacy and safety of the mRNA-1273 SARS-CoV-2 vaccineBaden, L. R.; El Sahly, H. M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S. A.; Rouphael, N.; Creech, C. B.; McGettigan, J.; Khetan, S.; Segall, N.; Solis, J.; Brosz, A.; Fierro, C.; Schwartz, H.; Neuzil, K.; Corey, L.; Gilbert, P.; Janes, H.; Follmann, D.; Marovich, M.; Mascola, J.; Polakowski, L.; Ledgerwood, J.; Graham, B. S.; Bennett, H.; Pajon, R.; Knightly, C.; Leav, B.; Deng, W.; Zhou, H.; Han, S.; Ivarsson, M.; Miller, J.; Zaks, T.New England Journal of Medicine (2021), 384 (5), 403-416CODEN: NEJMAG; ISSN:1533-4406. (Massachusetts Medical Society)Vaccines are needed to prevent coronavirus disease 2019 (Covid-19) and to protect persons who are at high risk for complications. The mRNA-1273 vaccine is a lipid nanoparticle-encapsulated mRNA-based vaccine that encodes the prefusion stabilized full-length spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes Covid-19. This phase 3 randomized, observer-blinded, placebo-controlled trial was conducted at 99 centers across the United States. Persons at high risk for SARS-CoV-2 infection or its complications were randomly assigned in a 1:1 ratio to receive two i.m. injections of mRNA-1273 (100μg) or placebo 28 days apart. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second injection in participants who had not previously been infected with SARS-CoV-2. The trial enrolled 30,420 volunteers who were randomly assigned in a 1:1 ratio to receive either vaccine or placebo (15,210 participants in each group). More than 96% of participants received both injections, and 2.2% had evidence (serol., virol., or both) of SARS-CoV-2 infection at baseline. Symptomatic Covid-19 illness was confirmed in 185 participants in the placebo group (56.5 per 1000 person-years; 95% confidence interval [CI], 48.7 to 65.3) and in 11 participants in the mRNA-1273 group (3.3 per 1000 person-years; 95% CI, 1.7 to 6.0); vaccine efficacy was 94.1% (95% CI, 89.3 to 96.8%; P<0.001). Efficacy was similar across key secondary analyses, including assessment 14 days after the first dose, analyses that included participants who had evidence of SARS-CoV-2 infection at baseline, and analyses in participants 65 years of age or older. Severe Covid-19 occurred in 30 participants, with one fatality; all 30 were in the placebo group. Moderate, transient reactogenicity after vaccination occurred more frequently in the mRNA-1273 group. Serious adverse events were rare, and the incidence was similar in the two groups. The mRNA-1273 vaccine showed 94.1% efficacy at preventing Covid-19 illness, including severe disease. Aside from transient local and systemic reactions, no safety concerns were identified.
- 20Kovyrshina, A. V.; Dolzhikova, I. V.; Grousova, D. M. A heterologous virus-vectored vaccine for prevention of middle east respiratory syndrome induces long protective immune response against MERS-CoV. Immunologiya 2020, 41, 135– 43, DOI: 10.33029/0206-4952-2020-41-2-135-143There is no corresponding record for this reference.
- 21Logunov, D. Y.; Dolzhikova, I. V.; Zubkova, O. V. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet 2020, 396, 887– 97, DOI: 10.1016/S0140-6736(20)31866-321https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsl2ktrfF&md5=68963e757a4869be9a28726203eacc8bSafety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from RussiaLogunov, Denis Y.; Dolzhikova, Inna V.; Zubkova, Olga V.; Tukhvatullin, Amir I.; Shcheblyakov, Dmitry V.; Dzharullaeva, Alina S.; Grousova, Daria M.; Erokhova, Alina S.; Kovyrshina, Anna V.; Botikov, Andrei G.; Izhaeva, Fatima M.; Popova, Olga; Ozharovskaya, Tatiana A.; Esmagambetov, Ilias B.; Favorskaya, Irina A.; Zrelkin, Denis I.; Voronina, Daria V.; Shcherbinin, Dmitry N.; Semikhin, Alexander S.; Simakova, Yana V.; Tokarskaya, Elizaveta A.; Lubenets, Nadezhda L.; Egorova, Daria A.; Shmarov, Maksim M.; Nikitenko, Natalia A.; Morozova, Lola F.; Smolyarchuk, Elena A.; Kryukov, Evgeny V.; Babira, Vladimir F.; Borisevich, Sergei V.; Naroditsky, Boris S.; Gintsburg, Alexander L.Lancet (2020), 396 (10255), 887-897CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)We developed a heterologous COVID-19 vaccine consisting of 2 components, a recombinant adenovirus type 26 (rAd26) vector and a recombinant adenovirus type 5 (rAd5) vector, both carrying the gene for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein (rAd26-S and rAd5-S). We aimed to assess the safety and immunogenicity of 2 formulations (frozen and lyophilized) of this vaccine. We did 2 open, non-randomized phase 1/2 studies at 2 hospitals in Russia. We enrolled healthy adult volunteers (men and women) aged 18-60 yr to both studies. In phase 1 of each study, we administered i.m. on day 0 either 1 dose of rAd26-S or 1 dose of rAd5-S and assessed the safety of the 2 components for 28 days. In phase 2 of the study, which began no earlier than 5 days after phase 1 vaccination, we administered i.m. a prime-boost vaccination, with rAd26-S given on day 0 and rAd5-S on day 21. Primary outcome measures were antigen-specific humoral immunity (SARS-CoV-2-specific antibodies measured by ELISA on days 0, 14, 21, 28, and 42) and safety (no. of participants with adverse events monitored throughout the study). Secondary outcome measures were antigen-specific cellular immunity (T-cell responses and interferon-γ concn.) and change in neutralizing antibodies (detected with a SARS-CoV-2 neutralization assay). These trials are registered with ClinicalTrials.gov, NCT04436471 and NCT04437875. Between June 18 and Aug 3, 2020, we enrolled 76 participants to the 2 studies (38 in each study). In each study, 9 volunteers received rAd26-S in phase 1, 9 received rAd5-S in phase 1, and 20 received rAd26-S and rAd5-S in phase 2. Both vaccine formulations were safe and well tolerated. The most common adverse events were pain at injection site (44 [58%]), hyperthermia (38 [50%]), headache (32 [42%]), asthenia (21 [28%]), and muscle and joint pain (18 [24%]). Most adverse events were mild and no serious adverse events were detected. All participants produced antibodies to SARS-CoV-2 glycoprotein. At day 42, receptor binding domain-specific IgG titers were 14 703 with the frozen formulation and 11,143 with the lyophilized formulation, and neutralizing antibodies were 49·25 with the frozen formulation and 45·95 with the lyophilized formulation, with a seroconversion rate of 100%. Cell-mediated responses were detected in all participants at day 28, with median cell proliferation of 2.5% CD4+ and 1.3% CD8+ with the frozen formulation, and a median cell proliferation of 1.3% CD4+ and 1.1% CD8+ with the lyophilized formulation. The heterologous rAd26 and rAd5 vector-based COVID-19 vaccine has a good safety profile and induced strong humoral and cellular immune responses in participants. Further investigation is needed of the effectiveness of this vaccine for prevention of COVID-19. Ministry of Health of the Russian Federation.
- 22Logunov, D. Y.; Dolzhikova, I. V.; Shcheblyakov, D. V.; Tukhvatulin, A. Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomised controlled phase 3 trial in Russia. Lancet 2021, 397, 671, DOI: 10.1016/S0140-6736(21)00234-822https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjslajtrs%253D&md5=94062efdc9d84abcfe49ebae871934a4Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomized controlled phase 3 trial in RussiaLogunov, Denis Y.; Dolzhikova, Inna V.; Shcheblyakov, Dmitry V.; Tukhvatulin, Amir I.; Zubkova, Olga V.; Dzharullaeva, Alina S.; Kovyrshina, Anna V.; Lubenets, Nadezhda L.; Grousova, Daria M.; Erokhova, Alina S.; Botikov, Andrei G.; Izhaeva, Fatima M.; Popova, Olga; Ozharovskaya, Tatiana A.; Esmagambetov, Ilias B.; Favorskaya, Irina A.; Zrelkin, Denis I.; Voronina, Daria V.; Shcherbinin, Dmitry N.; Semikhin, Alexander S.; Simakova, Yana V.; Tokarskaya, Elizaveta A.; Egorova, Daria A.; Shmarov, Maksim M.; Nikitenko, Natalia A.; Gushchin, Vladimir A.; Smolyarchuk, Elena A.; Zyryanov, Sergey K.; Borisevich, Sergei V.; Naroditsky, Boris S.; Gintsburg, Alexander L.Lancet (2021), 397 (10275), 671-681CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)A heterologous recombinant adenovirus (rAd)-based vaccine, Gam-COVID-Vac (Sputnik V), showed a good safety profile and induced strong humoral and cellular immune responses in participants in phase 1/2 clin. trials. Here, we report preliminary results on the efficacy and safety of Gam-COVID-Vac from the interim anal. of this phase 3 trial. We did a randomized, double-blind, placebo-controlled, phase 3 trial at 25 hospitals and polyclinics in Moscow, Russia. We included participants aged at least 18 years, with neg. SARS-CoV-2 PCR and IgG and IgM tests, no infectious diseases in the 14 days before enrollment, and no other vaccinations in the 30 days before enrollment. Participants were randomly assigned (3:1) to receive vaccine or placebo, with stratification by age group. Investigators, participants, and all study staff were masked to group assignment. The vaccine was administered (0·5 mL/dose) i.m. in a prime-boost regimen: a 21-day interval between the first dose (rAd26) and the second dose (rAd5), both vectors carrying the gene for the full-length SARS-CoV-2 glycoprotein S. The primary outcome was the proportion of participants with PCR-confirmed COVID-19 from day 21 after receiving the first dose. All analyses excluded participants with protocol violations: the primary outcome was assessed in participants who had received two doses of vaccine or placebo, serious adverse events were assessed in all participants who had received at least one dose at the time of database lock, and rare adverse events were assessed in all participants who had received two doses and for whom all available data were verified in the case report form at the time of database lock. The trial is registered at ClinicalTrials.gov (NCT04530396). Between Sept 7 and Nov 24, 2020, 21 977 adults were randomly assigned to the vaccine group (n=16 501) or the placebo group (n=5476). 19 866 received two doses of vaccine or placebo and were included in the primary outcome anal. From 21 days after the first dose of vaccine (the day of dose 2), 16 (0·1%) of 14 964 participants in the vaccine group and 62 (1·3%) of 4902 in the placebo group were confirmed to have COVID-19; vaccine efficacy was 91·6% (95% CI 85·6-95·2). Most reported adverse events were grade 1 (7485 [94·0%] of 7966 total events). 45 (0·3%) of 16 427 participants in the vaccine group and 23 (0·4%) of 5435 participants in the placebo group had serious adverse events; none were considered assocd. with vaccination, with confirmation from the independent data monitoring committee. Four deaths were reported during the study (three [<0·1%] of 16 427 participants in the vaccine group and one [<0·1%] of 5435 participants in the placebo group), none of which were considered related to the vaccine. This interim anal. of the phase 3 trial of Gam-COVID-Vac showed 91·6% efficacy against COVID-19 and was well tolerated in a large cohort. Moscow City Health Department, Russian Direct Investment Fund, Sberbank, and RUSAL.
- 23WHO. Transmission of SARS-CoV-2: implications for infection prevention precautions. https://www.who.int/news-room/commentaries/detail/transmission-of-sars-cov-2-implications-for-infection-prevention-precautions (accessed January 2020).There is no corresponding record for this reference.
- 24Centers for disease control and prevention. Interim clinical guidance for management of patients with confirmed coronavirus disease (COVID-19). https://www.cdc.gov/coronavirus/2019-ncov/hcp/ (accessed May 2020).There is no corresponding record for this reference.
- 25WHO. Target product profiles for COVID-19 vaccines , 2020. https://www.who.int/publications/m/item/who-target-product-profiles-for-covid-19-vaccines (accessed January 2020).There is no corresponding record for this reference.
- 26Amanat, F. K. SARS-CoV-2 vaccines: status report. Immunity 2020, 52, 583, DOI: 10.1016/j.immuni.2020.03.00726https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXmsVygs7k%253D&md5=63071e7666cff2dd50c48fc51cdf3c99SARS-CoV-2 Vaccines: Status ReportAmanat, Fatima; Krammer, FlorianImmunity (2020), 52 (4), 583-589CODEN: IUNIEH; ISSN:1074-7613. (Elsevier Inc.)A review. SARS-CoV-2, the causal agent of COVID-19, first emerged in late 2019 in China. It has since infected more than 870,000 individuals and caused more than 43,000 deaths globally. Here, we discuss therapeutic and prophylactic interventions for SARS-CoV-2 with a focus on vaccine development and its challenges. Vaccines are being rapidly developed but will likely come too late to affect the first wave of a potential pandemic. Nevertheless, crit. lessons can be learned for the development of vaccines against rapidly emerging viruses. Importantly, SARS-CoV-2 vaccines will be essential to reducing morbidity and mortality if the virus establishes itself in the population.
- 27Chen, W. H.; Strych, U.; Hotez, P. J.; Bottazzi, M. E. The SARS-CoV-2 vaccine pipeline: an overview. Curr. Trop Med. Rep. 2020, 7, 61, DOI: 10.1007/s40475-020-00201-627https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB383otFWmtg%253D%253D&md5=10552f461e781e5eb4567cd3c14060ecThe SARS-CoV-2 Vaccine Pipeline: an OverviewChen Wen-Hsiang; Strych Ulrich; Hotez Peter J; Bottazzi Maria ElenaCurrent tropical medicine reports (2020), 7 (2), 61-64 ISSN:2196-3045.Purpose of Review: The goal of this review is to provide a timely overview on efforts to develop a vaccine for the 2019 novel coronavirus SARS-CoV-2, the causative agent of coronavirus disease (COVID-19). Recent Findings: Previous research efforts to develop a severe acute respiratory syndrome coronavirus (SARS-CoV) vaccine in the years following the 2003 pandemic have opened the door for investigators to design vaccine concepts and approaches for the COVID-19 epidemic in China. Both SARS-CoV and SARS-CoV-2 exhibit a high degree of genetic similarity and bind to the same host cell ACE2 receptor. Based on previous experience with SARS-CoV vaccines, it is expected that all COVID-19 vaccines will require careful safety evaluations for immunopotentiation that could lead to increased infectivity or eosinophilic infiltration. Besides this, a COVID-19 vaccine target product profile must address vaccinating at-risk human populations including frontline healthcare workers, individuals over the age of 60, and those with underlying and debilitating chronic conditions. Among the vaccine technologies under evaluation are whole virus vaccines, recombinant protein subunit vaccines, and nucleic acid vaccines. Summary: Each current vaccine strategy has distinct advantages and disadvantages. Therefore, it is paramount that multiple strategies be advanced quickly and then evaluated for safety and efficacy. Ultimately, the safety studies to minimize undesired immunopotentiation will become the most significant bottleneck in terms of time.
- 28Leader, B.; Baca, Q. J.; Golan, D. E. Protein therapeutics: a summary and pharmacological classification. Nat. Rev. Drug Discovery 2008, 7, 21– 39, DOI: 10.1038/nrd239928https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1ygtQ%253D%253D&md5=761a9860e38c98295385641dda94b31fProtein therapeutics: a summary and pharmacological classificationLeader, Benjamin; Baca, Quentin J.; Golan, David E.Nature Reviews Drug Discovery (2008), 7 (1), 21-39CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Once a rarely used subset of medical treatments, protein therapeutics have increased dramatically in no. and frequency of use since the introduction of the first recombinant protein therapeutic - human insulin - 25 years ago. Protein therapeutics already have a significant role in almost every field of medicine, but this role is still only in its infancy. This article overviews some of the key characteristics of protein therapeutics, summarizes the more than 130 protein therapeutics used currently and suggests a new classification of these proteins according to their pharmacol. action.
- 29Suntharalingam, G.; Perry, M. R.; Ward, S.; Brett, S. J.; Panoskaltsis, N. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 2006, 355, 1018– 1028, DOI: 10.1056/NEJMoa06384229https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpsFKmtbc%253D&md5=e2d2ace0459e5f0d7472de79e68ae37eCytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412Suntharalingam, Ganesh; Perry, Meghan R.; Ward, Stephen; Brett, Stephen J.; Castello-Cortes, Andrew; Brunner, Michael D.; Panoskaltsis, NickiNew England Journal of Medicine (2006), 355 (10), 1018-1028CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)Six healthy young male volunteers at a contract research organization were enrolled in the first phase 1 clin. trial of TGN1412, a novel superagonist anti-CD28 monoclonal antibody that directly stimulates T cells. Within 90 min after receiving a single i.v. dose of the drug, all six volunteers had a systemic inflammatory response characterized by a rapid induction of proinflammatory cytokines and accompanied by headache, myalgias, nausea, diarrhea, erythema, vasodilatation, and hypotension. Within 12 to 16 h after infusion, they became critically ill, with pulmonary infiltrates and lung injury, renal failure, and disseminated intravascular coagulation. Severe and unexpected depletion of lymphocytes and monocytes occurred within 24 h after infusion. All six patients were transferred to the care of the authors at an intensive care unit at a public hospital, where they received intensive cardiopulmonary support (including dialysis), high-dose methylprednisolone, and an anti-interleukin-2 receptor antagonist antibody. Prolonged cardiovascular shock and acute respiratory distress syndrome developed in two patients, who required intensive organ support for 8 and 16 days. Despite evidence of the multiple cytokine-release syndrome, all six patients survived. Documentation of the clin. course occurring over the 30 days after infusion offers insight into the systemic inflammatory response syndrome in the absence of contaminating pathogens, endotoxin, or underlying disease.
- 30Wadman, M. London’s disastrous drug trial has serious side effects for research. Nature 2006, 440, 388– 389, DOI: 10.1038/440388a30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xis1Omt7c%253D&md5=2516c376286b52865e6371181688bf14London's disastrous drug trial has serious side effects for researchWadman, MeredithNature (London, United Kingdom) (2006), 440 (7083), 388-389CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)There is no expanded citation for this reference.
- 31Huck, B. R.; Kötzner, L.; Urbahns, K. Small molecules drive big improvements in immuno-oncology therapies. Angew. Chem., Int. Ed. 2018, 57, 4412– 4428, DOI: 10.1002/anie.20170781631https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjtlWqsbo%253D&md5=bef3a381977686b5bdb432a76364b066Small Molecules Drive Big Improvements in Immuno-Oncology TherapiesHuck, Bayard R.; Koetzner, Lisa; Urbahns, KlausAngewandte Chemie, International Edition (2018), 57 (16), 4412-4428CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Immunooncol. therapies have the potential to revolutionize the armamentarium of available cancer treatments. To further improve clin. response rates, researchers are looking for novel combination regimens, with checkpoint blockade being used as a backbone of the treatment. This Review highlights the significance of small mols. in this approach, which holds promise to provide increased benefit to cancer patients.
- 32Gadek, T. R.; Burdick, D. J.; McDowell, R. S.; Stanley, M. S. Generation of an LFA-1 antagonist by the transfer of the ICAM-1 immunoregulatory epitope to a small molecule. Science 2002, 295, 1086– 1089, DOI: 10.1126/science.295.5557.108632https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38Xht1Gqt7s%253D&md5=bbcb9a2fe2c245e038dd7ddb28462234Generation of an LFA-1 antagonist by the transfer of the ICAM-1 immunoregulatory epitope to a small moleculeGadek, T. R.; Burdick, D. J.; McDowell, R. S.; Stanley, M. S.; Marsters, J. C., Jr.; Paris, K. J.; Oare, D. A.; Reynolds, M. E.; Ladner, C.; Zioncheck, K. A.; Lee, W. P.; Gribling, P.; Dennis, M. S.; Skelton, N. J.; Tumas, D. B.; Clark, K. R.; Keating, S. M.; Beresini, M. H.; Tilley, J. W.; Presta, L. G.; Bodary, S. C.Science (Washington, DC, United States) (2002), 295 (5557), 1086-1089CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The protein-protein interaction between leukocyte functional antigen-1 (LFA-1) and intercellular adhesion mol.-1 (ICAM-1) is crit. to lymphocyte and immune system function. Here, we report on the transfer of the contiguous, nonlinear epitope of ICAM-1, responsible for its assocn. with LFA-1, to a small-mol. framework. These LFA-1 antagonists bound LFA-1, blocked binding of ICAM-1, and inhibited a mixed lymphocyte reaction (MLR) with potency significantly greater than that of cyclosporine A. Furthermore, in comparison to an antibody to LFA-1, they exhibited significant anti-inflammatory effects in vivo. These results demonstrate the utility of small-mol. mimics of nonlinear protein epitopes and the protein epitopes themselves as leads in the identification of novel pharmaceutical agents.
- 33Scott, D. E.; Bayly, A. R.; Abell, C.; Skidmore, J. Small molecules, big targets: drug discovery faces the protein-protein interaction challenge. Nat. Rev. Drug Discovery 2016, 15, 533– 550, DOI: 10.1038/nrd.2016.2933https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XlsFOgur4%253D&md5=9f0953567ae9338b8b36aa78b8160655Small molecules, big targets: drug discovery faces the protein-protein interaction challengeScott, Duncan E.; Bayly, Andrew R.; Abell, Chris; Skidmore, JohnNature Reviews Drug Discovery (2016), 15 (8), 533-550CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)Protein-protein interactions (PPIs) are of pivotal importance in the regulation of biol. systems and are consequently implicated in the development of disease states. Recent work has begun to show that, with the right tools, certain classes of PPI can yield to the efforts of medicinal chemists to develop inhibitors, and the first PPI inhibitors have reached clin. development. In this Review, we describe the research leading to these breakthroughs and highlight the existence of groups of structurally related PPIs within the PPI target class. For each of these groups, we use examples of successful discovery efforts to illustrate the research strategies that have proved most useful.
- 34Zhang, G.; Pomplun, S.; Loftis, A. R.; Loas, A.; Pentelute, B. L. Investigation of ACE2 N-terminal fragments binding to SARS-CoV-2 spike RBD. bioRxiv , June 17, 2020, ver. 1. DOI: 10.1101/2020.03.19.999318 .There is no corresponding record for this reference.
- 35Han, Y.; Král, P. Computational design of ACE2-based peptide inhibitors of SARS-CoV-2. ACS Nano 2020, 14, 5143– 5147, DOI: 10.1021/acsnano.0c0285735https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXntVKns7s%253D&md5=54088dfed751aff6a9e8dcec95bd25edComputational design of ACE2-based peptide inhibitors of SARS-CoV-2Han, Yanxiao; Kral, PetrACS Nano (2020), 14 (4), 5143-5147CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Peptide inhibitors against the SARS-CoV-2 coronavirus, currently causing a worldwide pandemic, are designed and simulated. The inhibitors are mostly formed by two sequential self-supporting α-helixes (bundle) extd. from the protease domain (PD) of angiotensin-converting enzyme 2 (ACE2), which bind to the SARS-CoV-2 receptor binding domains. Mol. dynamics simulations revealed that the α-helical peptides maintain their secondary structure and provide a highly specific and stable binding (blocking) to SARS-CoV-2. To provide a multivalent binding to the SARS-CoV-2 receptors, many such peptides could be attached to the surfaces of nanoparticle carriers. The proposed peptide inhibitors could provide simple and efficient therapeutics against the COVID-19 disease.
- 36Karoyan, P.; Vieillard, V.; Gómez-Morales, L. Human ACE2 peptide-mimics block SARS-CoV-2 pulmonary cells infection. Commun. Biol. 2021, 4, 197, DOI: 10.1038/s42003-021-01736-836https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktlajsro%253D&md5=4750200c747ebdaab86ff52b6cf0e66aHuman ACE2 peptide-mimics block SARS-CoV-2 pulmonary cells infectionKaroyan, Philippe; Vieillard, Vincent; Gomez-Morales, Luis; Odile, Estelle; Guihot, Amelie; Luyt, Charles-Edouard; Denis, Alexis; Grondin, Pascal; Lequin, OlivierCommunications Biology (2021), 4 (1), 197CODEN: CBOIDQ; ISSN:2399-3642. (Nature Research)In light of the recent accumulated knowledge on SARS-CoV-2 and its mode of human cells invasion, the binding of viral spike glycoprotein to human Angiotensin Converting Enzyme 2 (hACE2) receptor plays a central role in cell entry. We designed a series of peptides mimicking the N-terminal helix of hACE2 protein which contains most of the contacting residues at the binding site, exhibiting a high helical folding propensity in aq. soln. Our best peptide-mimics are able to block SARS-CoV-2 human pulmonary cell infection with an inhibitory concn. (IC50) in the nanomolar range upon binding to the virus spike protein with high affinity. These first-in-class blocking peptide mimics represent powerful tools that might be used in prophylactic and therapeutic approaches to fight the coronavirus disease 2019 (COVID-19).
- 37Curreli, F.; Victor, S. M. B.; Ahmed, S.; Drelich, A. Stapled peptides based on human angiotensin-converting enzyme 2 (ACE2) potently inhibit SARS-CoV-2 infection in vitro. mBio 2020, 11, e02451-20 DOI: 10.1128/mBio.02451-20There is no corresponding record for this reference.
- 38Tai, W. Identification of SARS-CoV RBD-targeting monoclonal antibodies with cross-reactive or neutralizing activity against SARS-CoV-2. Antiviral Res. 2020, 179, 104820, DOI: 10.1016/j.antiviral.2020.10482038https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXpvFansb8%253D&md5=3418b1f745d3d23944bb6acef604a99dIdentification of SARS-CoV RBD-targeting monoclonal antibodies with cross-reactive or neutralizing activity against SARS-CoV-2Tai, Wanbo; Zhang, Xiujuan; He, Yuxian; Jiang, Shibo; Du, LanyingAntiviral Research (2020), 179 (), 104820CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)SARS-CoV-2-caused COVID-19 cases are growing globally, calling for developing effective therapeutics to control the current pandemic. SARS-CoV-2 and SARS-CoV recognize angiotensin-converting enzyme 2 (ACE2) receptor via the receptor-binding domain (RBD). Here, we identified six SARS-CoV RBD-specific neutralizing monoclonal antibodies (nAbs) that cross-reacted with SARS-CoV-2 RBD, two of which, 18F3 and 7B11, neutralized SARS-CoV-2 infection. 18F3 recognized conserved epitopes on SARS-CoV and SARS-CoV-2 RBDs, whereas 7B11 recognized epitopes on SARS-CoV RBD not fully conserved in SARS-CoV-2 RBD. The 18F3-recognizing epitopes on RBD did not overlap with the ACE2-binding sites, whereas those recognized by 7B11 were close to the ACE2-binding sites, explaining why 7B11 could, but 18F3 could not, block SARS-CoV or SARS-CoV-2 RBD binding to ACE2 receptor. Our study provides an alternative approach to prevent SARS-CoV-2 infection using anti-SARS-CoV nAbs.
- 39De Vries, R. D.; Schmitz, K. S.; Bovier, F. T. Intranasal fusion inhibitory lipopeptide prevents direct contact SARS-CoV-2 transmission in ferrets. bioRxiv , November 5, 2020, ver. 1. DOI: 10.1101/2020.11.04.361154 .There is no corresponding record for this reference.
- 40Tai, W.; He, L.; Zhang, X.; Pu, J.; Voronin, D.; Jiang, S.; Zhou, Y.; Du, L. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell. Mol. Immunol. 2020, 17, 613– 620, DOI: 10.1038/s41423-020-0400-440https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXlt1Chsrw%253D&md5=87bc49d070c84e78b01230518aaa465aCharacterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccineTai, Wanbo; He, Lei; Zhang, Xiujuan; Pu, Jing; Voronin, Denis; Jiang, Shibo; Zhou, Yusen; Du, LanyingCellular & Molecular Immunology (2020), 17 (6), 613-620CODEN: CMIEAO; ISSN:1672-7681. (Nature Research)The outbreak of Coronavirus Disease 2019 (COVID-19) has posed a serious threat to global public health, calling for the development of safe and effective prophylactics and therapeutics against infection of its causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as 2019 novel coronavirus (2019-nCoV). The CoV spike (S) protein plays the most important roles in viral attachment, fusion and entry, and serves as a target for development of antibodies, entry inhibitors and vaccines. Here, we identified the receptor-binding domain (RBD) in SARS-CoV-2 S protein and found that the RBD protein bound strongly to human and bat angiotensin-converting enzyme 2 (ACE2) receptors. SARS-CoV-2 RBD exhibited significantly higher binding affinity to ACE2 receptor than SARS-CoV RBD and could block the binding and, hence, attachment of SARS-CoV-2 RBD and SARS-CoV RBD to ACE2-expressing cells, thus inhibiting their infection to host cells. SARS-CoV RBD-specific antibodies could cross-react with SARS-CoV-2 RBD protein, and SARS-CoV RBD-induced antisera could cross-neutralize SARS-CoV-2, suggesting the potential to develop SARS-CoV RBD-based vaccines for prevention of SARS-CoV-2 and SARS-CoV infection.
- 41Sun, C.; Chen, L.; Yang, J. SARS-CoV-2 and SARS-CoV spike-RBD structure and receptor binding comparison and potential implications on neutralizing antibody and vaccine development. bioRxiv , February 20, 2020, ver. 1. DOI: 10.1101/2020.02.16.951723 .There is no corresponding record for this reference.
- 42Xia, S.; Yan, L.; Xu, W.; Agrawal, A. S.; Algaissi, A.; Tseng, C. K.; Wang, Q.; Du, L.; Tan, W.; Wilson, I. A.; Jiang, S.; Yang, B.; Lu, L. A pan-coronavirus fusion inhibitor targeting the HR1mdomain of human coronavirus spike. Sci. Adv. 2019, 5, eaav4580 DOI: 10.1126/sciadv.aav4580There is no corresponding record for this reference.
- 43Rathod, S. B.; Prajapati, P. B.; Punjabi, L. B.; Prajapati, K. N.; Chauhan, N.; Mansuri, M. F. Peptide modelling and screening against human ACE2 and spike glycoprotein RBD of SARS CoV 2. In Silico Pharmacol. 2020, 8, 3, DOI: 10.1007/s40203-020-00055-w43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s3kt1aqsQ%253D%253D&md5=41a5887a9cbbf2a0fa7e9cd70804aceaPeptide modelling and screening against human ACE2 and spike glycoprotein RBD of SARS-CoV-2Rathod Shravan B; Prajapati Pravin B; Prajapati Kuntal N; Punjabi Lata B; Chauhan Neha; Mansuri Mohmedyasin FIn silico pharmacology (2020), 8 (1), 3 ISSN:2193-9616.Outbreak of Coronavirus Disease 2019 (COVID-19) has become a great challenge for scientific community globally. Virus enters cell through spike glycoprotein fusion with ACE2 (Angiotensin-Converting Enzyme 2) human receptor. Hence, spike glycoprotein of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a potential target for diagnostics, vaccines, and antibodies. Also, virus entry can be prevented by blocking ACE2 thus, ACE2 can be considered potential target for therapeutics. As being highly specific, safe and efficacious, peptides hold their place in therapeutics. In present study, we retrieved sequence of 70 peptides from Antiviral Peptide Database (AVPdb), modelled them using 3D structure predicting web tool and docked them with receptor binding domain (RBD) of spike protein and human host receptor ACE2 using peptide-protein docking. It was observed that peptides have more affinity towards ACE2 in comparison with spike RBD. Interestingly it was noticed that most of the peptides bind to RBM (residue binding motif) which is responsible for ACE2 binding at the interface of RBD while, for ACE2, peptides prefer to bind the core cavity rather than RBD binding interface. To further investigate how peptides at the interface of RBD or ACE2 alter the binding between RBD and ACE2, protein-protein docking of RBD and ACE2 with and without peptides was performed. Peptides, AVP0671 at RBD and AVP1244 at ACE2 interfaces significantly reduce the binding affinity and change the orientation of RBD and ACE2 binding. This finding suggests that peptides can be used as a drug to inhibit virus entry in cells to stop COVID-19 pandemic in the future after experimental evidences.
- 44Watson, A.; Ferreira, L.; Hwang, P.; Xu, J.; Stroud, R. Peptide antidotes to SARS-CoV-2 (COVID-19). bioRxiv , August 6, 2020, ver. 1. DOI: 10.1101/2020.08.06.238915 .There is no corresponding record for this reference.
- 45Singh, A.; Thakur, M.; Sharma, L. K.; Chandra, K. Designing a multi epitope peptide based vaccine against SARS CoV 2. Sci. Rep. 2020, 10, 16219, DOI: 10.1038/s41598-020-73371-y45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvF2ksbrM&md5=47ec0379065f691b7fed33b5216556c5Designing a multi-epitope peptide based vaccine against SARS-CoV-2Singh, Abhishek; Thakur, Mukesh; Sharma, Lalit Kumar; Chandra, KailashScientific Reports (2020), 10 (1), 16219CODEN: SRCEC3; ISSN:2045-2322. (Nature Research)COVID-19 pandemic has resulted in 16,114,449 cases with 646,641 deaths from the 217 countries, or territories as on July 27th 2020. Due to multifaceted issues and challenges in the implementation of the safety and preventive measures, inconsistent coordination between societies-governments and most importantly lack of specific vaccine to SARS-CoV-2, the spread of the virus that initially emerged at Wuhan is still uprising after taking a heavy toll on human life. In the present study, we mapped immunogenic epitopes present on the four structural proteins of SARS-CoV-2 and we designed a multi-epitope peptide based vaccine that, demonstrated a high immunogenic response with a vast application on world's human population. On codon optimization and in-silico cloning, we found that candidate vaccine showed high expression in E. coli and immune simulation resulted in inducing a high level of both B-cell and T-cell mediated immunity. The results predicted that exposure of vaccine by administrating three injections significantly subsidized the antigen growth in the system. The proposed candidate vaccine found promising by yielding desired results and hence, should be validated by practical experimentations for its functioning and efficacy to neutralize SARS-CoV-2.
- 46Molina, R.; Oliva, B.; Fernandez-Fuentes, N. A collection of designed peptides to target SARS-Cov-2 – ACE2 interaction: PepI-Covid19 database. bioRxiv , April 29, 2020, ver. 1. DOI: 10.1101/2020.04.28.051789 .There is no corresponding record for this reference.
- 47Zhao, H.; To, K. K. W.; Sze, K. H.; Yung, T. T.; Bian, M.; Lam, H.; Yeung, M. L.; Li, C.; Chu, H.; Yuen, K. Y. A broad-spectrum virus- and host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV-2. Nat. Commun. 2020, 11, 4252, DOI: 10.1038/s41467-020-17986-947https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs12rurbM&md5=9d48f2626060fca048216f3a6cfa840dA broad-spectrum virus- and host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV-2Zhao, Hanjun; To, Kelvin K. W.; Sze, Kong-Hung; Yung, Timothy Tin-Mong; Bian, Mingjie; Lam, Hoiyan; Yeung, Man Lung; Li, Cun; Chu, Hin; Yuen, Kwok-YungNature Communications (2020), 11 (1), 4252CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)The 2019 novel respiratory virus (SARS-CoV-2) causes COVID-19 with rapid global socioeconomic disruptions and disease burden to healthcare. The COVID-19 and previous emerging virus outbreaks highlight the urgent need for broad-spectrum antivirals. A defensin-like peptide P9R exhibited potent antiviral activity against pH-dependent viruses that require endosomal acidification for virus infection, including the enveloped pandemic A(H1N1)pdm09 virus, avian influenza A(H7N9) virus, coronaviruses (SARS-CoV-2, MERS-CoV and SARS-CoV), and the non-enveloped rhinovirus. P9R can significantly protect mice from lethal challenge by A(H1N1)pdm09 virus and shows low possibility to cause drug-resistant virus. Mechanistic studies indicate that the antiviral activity of P9R depends on the direct binding to viruses and the inhibition of virus-host endosomal acidification, which provides a proof of concept that virus-binding alk. peptides can broadly inhibit pH-dependent viruses. These results suggest that the dual-functional virus- and host-targeting P9R can be a promising candidate for combating pH-dependent respiratory viruses.
- 48Han, D. P.; Penn-Nicholson, A.; Cho, M. W. Identification of critical determinants on ACE2 for SARS-CoV entry and development of a potent entry inhibitor. Virology 2006, 350 (1), 15– 25, DOI: 10.1016/j.virol.2006.01.02948https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XlsFKrt74%253D&md5=1db622935cd76ac9a4a74cf6f6b186d5Identification of critical determinants on ACE2 for SARS-CoV entry and development of a potent entry inhibitorHan, Dong P.; Penn-Nicholson, Adam; Cho, Michael W.Virology (2006), 350 (1), 15-25CODEN: VIRLAX; ISSN:0042-6822. (Elsevier)Severe acute respiratory syndrome (SARS) is caused by a novel coronavirus, SARS-CoV. Virus entry into cells is mediated through interactions between spike (S) glycoprotein and angiotensin-converting enzyme 2 (ACE2). Alanine scanning mutagenesis anal. was performed to identify determinants on ACE2 crit. for SARS-CoV infection. Results indicated that charged amino acids between residues 22 and 57 were important, K26 and D30, in particular. Peptides representing various regions of ACE2 crit. for virus infection were chem. synthesized and evaluated for antiviral activity. Two peptides (a.a. 22-44 and 22-57) exhibited a modest antiviral activity with IC50 of about 50 μM and 6 μM, resp. One peptide comprised of two discontinuous segments of ACE2 (a.a. 22-44 and 351-357) artificially linked together by glycine, exhibited a potent antiviral activity with IC50 of about 0.1 μM. This novel peptide is a promising candidate as a therapeutic agent against this deadly emerging pathogen.
- 49Shang, J.; Ye, G.; Shi, K.; Wan, Y.; Luo, C.; Aihara, H.; Geng, Q.; Auerbach, A.; Li, F. Structural basis of receptor recognition by SARS-CoV-2. Nature 2020, 581, 221– 224, DOI: 10.1038/s41586-020-2179-y49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtbs%253D&md5=33bc9151641b2adcfb0dbf446621a1dcStructural basis of receptor recognition by SARS-CoV-2Shang, Jian; Ye, Gang; Shi, Ke; Wan, Yushun; Luo, Chuming; Aihara, Hideki; Geng, Qibin; Auerbach, Ashley; Li, FangNature (London, United Kingdom) (2020), 581 (7807), 221-224CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A novel severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) recently emerged and is rapidly spreading in humans, causing COVID-191,2. A key to tackling this pandemic is to understand the receptor recognition mechanism of the virus, which regulates its infectivity, pathogenesis and host range. SARS-CoV-2 and SARS-CoV recognize the same receptor-angiotensin-converting enzyme 2 (ACE2)-in humans3,4. Here we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 (engineered to facilitate crystn.) in complex with ACE2. In comparison with the SARS-CoV RBD, an ACE2-binding ridge in SARS-CoV-2 RBD has a more compact conformation; moreover, several residue changes in the SARS-CoV-2 RBD stabilize two virus-binding hotspots at the RBD-ACE2 interface. These structural features of SARS-CoV-2 RBD increase its ACE2-binding affinity. Addnl., we show that RaTG13, a bat coronavirus that is closely related to SARS-CoV-2, also uses human ACE2 as its receptor. The differences among SARS-CoV-2, SARS-CoV and RaTG13 in ACE2 recognition shed light on the potential animal-to-human transmission of SARS-CoV-2. This study provides guidance for intervention strategies that target receptor recognition by SARS-CoV-2.
- 50Zhou, T.; Tsybovsky, Y.; Gorman, J.; Rapp, M.; Cerutti, G. Cryo-EM structures of SARS-CoV-2 spike without and with ACE2 reveal a pH-dependent switch to mediate endosomal positioning of receptor-binding domains. Cell Host Microbe 2020, 28 (6), 867– 879, DOI: 10.1016/j.chom.2020.11.00450https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisVagtrjF&md5=e2cdd111aa8d7ec2ec774c62c26e6710Cryo-EM Structures of SARS-CoV-2 Spike without and with ACE2 Reveal a pH-Dependent Switch to Mediate Endosomal Positioning of Receptor-Binding DomainsZhou, Tongqing; Tsybovsky, Yaroslav; Gorman, Jason; Rapp, Micah; Cerutti, Gabriele; Chuang, Gwo-Yu; Katsamba, Phinikoula S.; Sampson, Jared M.; Schon, Arne; Bimela, Jude; Boyington, Jeffrey C.; Nazzari, Alexandra; Olia, Adam S.; Shi, Wei; Sastry, Mallika; Stephens, Tyler; Stuckey, Jonathan; Teng, I-Ting; Wang, Pengfei; Wang, Shuishu; Zhang, Baoshan; Friesner, Richard A.; Ho, David D.; Mascola, John R.; Shapiro, Lawrence; Kwong, Peter D.Cell Host & Microbe (2020), 28 (6), 867-879.e5CODEN: CHMECB; ISSN:1931-3128. (Elsevier Inc.)The SARS-CoV-2 spike employs mobile receptor-binding domains (RBDs) to engage the human ACE2 receptor and to facilitate virus entry, which can occur through low-pH-endosomal pathways. To understand how ACE2 binding and low pH affect spike conformation, we detd. cryo-electron microscopy structures-at serol. and endosomal pH-delineating spike recognition of up to three ACE2 mols. RBDs freely adopted "up" conformations required for ACE2 interaction, primarily through RBD movement combined with smaller alterations in neighboring domains. In the absence of ACE2, single-RBD-up conformations dominated at pH 5.5, resolving into a solitary all-down conformation at lower pH. Notably, a pH-dependent refolding region (residues 824-858) at the spike-interdomain interface displayed dramatic structural rearrangements and mediated RBD positioning through coordinated movements of the entire trimer apex. These structures provide a foundation for understanding prefusion-spike mechanics governing endosomal entry; we suggest that the low pH all-down conformation potentially facilitates immune evasion from RBD-up binding antibody.
- 51Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; Wang, X. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 2020, 581, 215– 220, DOI: 10.1038/s41586-020-2180-551https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXoslOqtL8%253D&md5=279c60143e8e5eb505457e0778baa8efStructure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptorLan, Jun; Ge, Jiwan; Yu, Jinfang; Shan, Sisi; Zhou, Huan; Fan, Shilong; Zhang, Qi; Shi, Xuanling; Wang, Qisheng; Zhang, Linqi; Wang, XinquanNature (London, United Kingdom) (2020), 581 (7807), 215-220CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new and highly pathogenic coronavirus (severe acute respiratory syndrome coronavirus-2, SARS-CoV-2) caused an outbreak in Wuhan city, Hubei province, China, starting from Dec. 2019 that quickly spread nationwide and to other countries around the world1-3. Here, to better understand the initial step of infection at an at. level, we detd. the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 bound to the cell receptor ACE2. The overall ACE2-binding mode of the SARS-CoV-2 RBD is nearly identical to that of the SARS-CoV RBD, which also uses ACE2 as the cell receptor4. Structural anal. identified residues in the SARS-CoV-2 RBD that are essential for ACE2 binding, the majority of which either are highly conserved or share similar side chain properties with those in the SARS-CoV RBD. Such similarity in structure and sequence strongly indicate convergent evolution between the SARS-CoV-2 and SARS-CoV RBDs for improved binding to ACE2, although SARS-CoV-2 does not cluster within SARS and SARS-related coronaviruses1-3,5. The epitopes of two SARS-CoV antibodies that target the RBD are also analyzed for binding to the SARS-CoV-2 RBD, providing insights into the future identification of cross-reactive antibodies.
- 52Sadremomtaz, A.; Ali, A. M.; Jouyandeh, F.; Balalaie, S.; Navari, R. Molecular docking, synthesis and biological evaluation of vascular endothelial growth factor (VEGF) B based peptide as antiangiogenic agent targeting the second domain of the vascular endothelial growth factor receptor 1 (VEGFR1D2) for anticancer application. Sig. Transduct. Target Ther. 2020, 5, 76, DOI: 10.1038/s41392-020-0177-zThere is no corresponding record for this reference.
- 53Sadremomtaz, A.; Mansouri, K.; Alemzadeh, G.; Safa, M.; Rastaghi, A. E.; Asghari, S. M. Dual blockade of VEGFR1 and VEGFR2 by a novel peptide abrogates VEGF driven angiogenesis, tumor growth, and metastasis through PI3K/ AKT and MAPK/ERK1/2 pathway. Biochim. Biophys. Acta, Gen. Subj. 2018, 1862, 2688– 2700, DOI: 10.1016/j.bbagen.2018.08.01353https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhs1Wnu7vJ&md5=ce96e6bf828c9f08f06949e6bdb3a295Dual blockade of VEGFR1 and VEGFR2 by a novel peptide abrogates VEGF-driven angiogenesis, tumor growth, and metastasis through PI3K/AKT and MAPK/ERK1/2 pathwaySadremomtaz, Afsaneh; Mansouri, Kamran; Alemzadeh, Golnaz; Safa, Majid; Rastaghi, Ahmadreza Esmaeili; Asghari, S. MohsenBiochimica et Biophysica Acta, General Subjects (2018), 1862 (12), 2688-2700CODEN: BBGSB3; ISSN:0304-4165. (Elsevier B.V.)Neutralization of vascular endothelial growth factor receptor 1 (VEGFR1) and/or VEGFR2 is a widely used means of inhibiting tumor angiogenesis. Based on the complex X-ray structures of VEGFA/VEGFR1, VEGFA/VEGFR2, and VEGFB/VEGFR1, a peptide (referred to as VGB) was designed to simultaneously bind to VEGFR1 and VEGFR2, and binding, antiangiogenic and antitumor properties of the peptide was investigated in vitro. VGB bound to both VEGFR1 and VEGFR2 in human umbilical vein endothelial cells (HUVECs), and inhibited the proliferation of HUVECs, 4 T1 mammary carcinoma tumor (MCT) cells in human umbilical vein endothelial cells (HUVECs) and 4T1 mammary carcinoma tumor (MCT) cells, and inhibited the proliferation of HUVE, 4T1 MCT, and U87 glioblastoma cells. Through abrogation of AKT and ERK1/2 phosphorylation, VEGFA-stimulated proliferation, migration, and two- and three-dimensional tube formation in HUVECs were inhibited more potently by VGB than by bevacizumab. In a murine 4 T1 MCT model, VGB strongly inhibited tumor growth without causing wt. loss, accompanied by inhibition of AKT and ERK1/2 phosphorylation, a significant decrease in tumor cell proliferation (Ki-67 expression), angiogenesis (CD31 and CD34 expression), an increase in apoptosis index (increased TUNEL staining and p53 expression and decreased Bcl-2 expression), and the suppression of systematic spreading of the tumor (reduced NF-κB and MMP-9 and increased E-cadherin expression). The dual specificity of VGB for VEGFR1 and VEGFR2, through which the PI3K/AKT and MAPK/ERK1/2 signaling pathways can be abrogated and, subsequently, angiogenesis, tumor growth, and metastasis are inhibited. This study demonstrated that simultaneous blockade of VEGFR1 and VEGFR2 downstream cascades is an effective means for treatment of various angiogenic disorders, esp. cancer.
- 54Sadremomtaz, A.; Kobarfard, F.; Mansouri, K.; Mirzanejad, L.; Asghari, S. M. Suppression of migratory and metastatic pathways via blocking VEGFR1 and VEGFR2. J. Recept. Signal Transduction Res. 2018, 38, 432– 441, DOI: 10.1080/10799893.2019.156778554https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cbpvFartg%253D%253D&md5=e67668413d5631e86f813c3d8d1baed2Suppression of migratory and metastatic pathways via blocking VEGFR1 and VEGFR2Sadremomtaz Afsaneh; Mirzanejad Laleh; Asghari S Mohsen; Kobarfard Farzad; Mansouri KamranJournal of receptor and signal transduction research (2018), 38 (5-6), 432-441 ISSN:.BACKGROUND: Vascular endothelial growth factor (VEGF) A and B are endothelial cell mitogens whose ligation to VEGFR1/VEGFR2 drives tumor angiogenesis and metastasis, and epithelial-mesenchymal transition (EMT). Blockade of these signaling axes could be obtained by disturbing the interactions between VEGFA and/or VEGFB with VEGFR1 and/or VEGFR2. METHODS: A 14-mer peptide (VGB) that recognizes both VEGFR1 and VEGFR2 were investigated for its inhibitory effects on the VEGF-induced proliferation and migration using MTT and scratch assay, respectively. Downstream signaling pathways were also assessed by quantitative estimation of gene and protein expression using real-time PCR and immunohistochemistry (IHC). RESULTS: We investigated the inhibitory effects of VGB on downstream mediators of metastasis, including epithelial-cadherin (E-cadherin), matrix metalloprotease-9 (MMP-9), cancer myelocytomatosis (c-Myc), and nuclear factor-κβ (NF-κβ), and migration, comprising focal adhesion kinase (FAK) and its substrate Paxilin. VGB inhibited the VEGF-induced proliferation of human umbilical vein endothelial cells (HUVECs), 4T1 and U87 cells in a time- and dose-dependent manner and migration of HUVECs. Based on IHC analyses, treatment of 4T1 mammary carcinoma tumor with VGB led to the suppression of p-AKT, p-ERK1/2, MMP-9, NF-κβ, and activation of E-cadherin compared with PBS-treated controls. Moreover, quantitative real-time PCR analyses of VGB-treated tumors revealed the reduced expression level of FAK, Paxilin, NF-κβ, MMP-9, c-Myc, and increased expression level of E-cadherin compared to PBS-treated controls. CONCLUSIONS: Our results demonstrated that simultaneous blockade of VEGFR1/VEGFR2 is an effective strategy to fight solid tumors by targeting a wider range of mediators involved in tumor angiogenesis, growth, and metastasis.
- 55Abraham, M. J.; Murtola, T.; Schulz, R.; Pall, S.; Smith, J. C.; Hess, B.; Lindahl, E. GROMACS: High performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX. 2015, 1 (2), 19– 25, DOI: 10.1016/j.softx.2015.06.001There is no corresponding record for this reference.
- 56Jo, S.; Kim, T.; Iyer, V. G.; Im, W. CHARMM-GUI: a web-based graphical user interface for CHARMM. J. Comput. Chem. 2008, 29, 1859– 1865, DOI: 10.1002/jcc.2094556https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXosVKksbc%253D&md5=112a3dd61d792b040f9f716b32220d7eCHARMM-GUI: a web-based graphical user interface for CHARMMJo, Sunhwan; Kim, Taehoon; Iyer, Vidyashankara G.; Im, WonpilJournal of Computational Chemistry (2008), 29 (11), 1859-1865CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)CHARMM is an academic research program used widely for macromol. mechanics and dynamics with versatile anal. and manipulation tools of at. coordinates and dynamics trajectories. CHARMM-GUI, http://www.charmm-gui.org, has been developed to provide a web-based graphical user interface to generate various input files and mol. systems to facilitate and standardize the usage of common and advanced simulation techniques in CHARMM. The web environment provides an ideal platform to build and validate a mol. model system in an interactive fashion such that, if a problem is found through visual inspection, one can go back to the previous setup and regenerate the whole system again. In this article, we describe the currently available functional modules of CHARMM-GUI Input Generator that form a basis for the advanced simulation techniques. Future directions of the CHARMM-GUI development project are also discussed briefly together with other features in the CHARMM-GUI website, such as Archive and Movie Gallery.
- 57Gowers, R. J.; Linke, M.; Barnoud, J.; Reddy, T. J. E.; Melo, M. N.; Seyler, S. MDanalysis: a python package for the rapid analysis of molecular dynamics simulations. Proceedings of the 15th python in science conference 2016, 98– 105, DOI: 10.25080/Majora-629e541a-00eThere is no corresponding record for this reference.
- 58Salentin, S.; Schreiber, S.; Haupt, V. J.; Adasme, M. F.; Schroeder, M. PLIP: fully automated protein–ligand interaction profiler. Nucleic Acids Res. 2015, 43, W443– W447, DOI: 10.1093/nar/gkv31558https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVymtbrI&md5=4878881a7bd715e5585682876e102e65PLIP: fully automated protein-ligand interaction profilerSalentin, Sebastian; Schreiber, Sven; Haupt, V. Joachim; Adasme, Melissa F.; Schroeder, MichaelNucleic Acids Research (2015), 43 (W1), W443-W447CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The characterization of interactions in protein-ligand complexes is essential for research in structural bioinformatics, drug discovery and biol. However, comprehensive tools are not freely available to the research community. Here, we present the protein-ligand interaction profiler (PLIP), a novel web service for fully automated detection and visualization of relevant non-covalent protein-ligand contacts in 3D structures, freely available at projects.biotec.tu-dresden.de/plip-web. The input is either a Protein Data Bank structure, a protein or ligand name, or a custom protein-ligand complex (e.g. from docking). In contrast to other tools, the rule-based PLIP algorithm does not require any structure prepn. It returns a list of detected interactions on single atom level, covering seven interaction types (hydrogen bonds, hydrophobic contacts, pi-stacking, pi-cation interactions, salt bridges, water bridges and halogen bonds). PLIP stands out by offering publication-ready images, PyMOL sesions files to generate custom images and parsable result files to facilitate successive data processing. The full python source code is available for download on the website. PLIP's command-line mode allows for high-throughput interaction profiling.
- 59Kayikci, M.; Venkatakrishnan, A. J.; Scott-Brown, J. Visualization and analysis of non-covalent contacts using the protein contacts atlas. Nat. Struct. Mol. Biol. 2018, 25, 185– 194, DOI: 10.1038/s41594-017-0019-z59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlCqu7rF&md5=b72bfcbc3e4e573920fb9f1f0403de88Visualization and analysis of non-covalent contacts using the Protein Contacts AtlasKayikci, Melis; Venkatakrishnan, A. J.; Scott-Brown, James; Ravarani, Charles N. J.; Flock, Tilman; Babu, M. MadanNature Structural & Molecular Biology (2018), 25 (2), 185-194CODEN: NSMBCU; ISSN:1545-9993. (Nature Research)Visualizations of biomol. structures empower us to gain insights into biol. functions, generate testable hypotheses, and communicate biol. concepts. Typical visualizations (such as ball and stick) primarily depict covalent bonds. In contrast, non-covalent contacts between atoms, which govern normal physiol., pathogenesis, and drug action, are seldom visualized. We present the Protein Contacts Atlas, an interactive resource of non-covalent contacts from over 100,000 PDB crystal structures. We developed multiple representations for visualization and anal. of non-covalent contacts at different scales of organization: atoms, residues, secondary structure, subunits, and entire complexes. The Protein Contacts Atlas enables researchers from different disciplines to investigate diverse questions in the framework of non-covalent contacts, including the interpretation of allostery, disease mutations and polymorphisms, by exploring individual subunits, interfaces, and protein-ligand contacts and by mapping external information. The Protein Contacts Atlas is available at http://www.mrc-lmb.cam.ac.uk/pca/ and also through PDBe.
- 60Azad, T.; Singaravelu, R.; Taha, Z.; Boulton, S. Nanoluciferase complementation-based biosensor reveals the importance of N- linked glycosylation of SARS-CoV-2 spike for viral entry. Mol. Ther. 2021, 29, 1984, DOI: 10.1016/j.ymthe.2021.02.00760https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXltV2hsr0%253D&md5=f96a869c49349aef8a2946ee1d0e5090Nanoluciferase complementation-based bioreporter reveals the importance of N-linked glycosylation of SARS-CoV-2 S for viral entryAzad, Taha; Singaravelu, Ragunath; Taha, Zaid; Jamieson, Taylor R.; Boulton, Stephen; Crupi, Mathieu J. F.; Martin, Nikolas T.; Brown, Emily E. F.; Poutou, Joanna; Ghahremani, Mina; Pelin, Adrian; Nouri, Kazem; Rezaei, Reza; Marshall, Christopher Boyd; Enomoto, Masahiro; Arulanandam, Rozanne; Alluqmani, Nouf; Samson, Reuben; Gingras, Anne-Claude; Cameron, D. William; Greer, Peter A.; Ilkow, Carolina S.; Diallo, Jean-Simon; Bell, John C.Molecular Therapy (2021), 29 (6), 1984-2000CODEN: MTOHCK; ISSN:1525-0024. (Cell Press)The ongoing COVID-19 pandemic has highlighted the immediate need for the development of antiviral therapeutics targeting different stages of the SARS-CoV-2 life cycle. We developed a bioluminescence-based bioreporter to interrogate the interaction between the SARS-CoV-2 viral spike (S) protein and its host entry receptor, angiotensin-converting enzyme 2 (ACE2). The bioreporter assay is based on a nanoluciferase complementation reporter, composed of two subunits, large BiT and small BiT, fused to the S receptor-binding domain (RBD) of the SARS-CoV-2 S protein and ACE2 ectodomain, resp. Using this bioreporter, we uncovered crit. host and viral determinants of the interaction, including a role for glycosylation of asparagine residues within the RBD in mediating successful viral entry. We also demonstrate the importance of N-linked glycosylation to the RBD's antigenicity and immunogenicity. Our study demonstrates the versatility of our bioreporter in mapping key residues mediating viral entry as well as screening inhibitors of the ACE2-RBD interaction. Our findings point toward targeting RBD glycosylation for therapeutic and vaccine strategies against SARS-CoV-2.
- 61Spiliotopoulos, D.; Kastritis, P. L.; Melquiond, A. S.; Bonvin, A. M.; Musco, G.; Rocchia, W.; Spitaleri, A. dMM-PBSA: A new HADDOCK scoring function for protein-peptide docking. Front Mol. Biosci. 2016, 3, 46, DOI: 10.3389/fmolb.2016.0004661https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhvFSitL%252FO&md5=01d6e4223bf1a6329e25ec6ad7802cd0dMM-PBSA: a new HADDOCK scoring function for protein-peptide dockingSpiliotopoulos, Dimitrios; Kastritis, Panagiotis L.; Melquiond, Adrien S. J.; Bonvin, Alexandre M. J. J.; Musco, Giovanna; Rocchia, Walter; Spitaleri, AndreaFrontiers in Molecular Biosciences (2016), 3 (), 46/1-46/13CODEN: FMBRBS; ISSN:2296-889X. (Frontiers Media S.A.)Mol.-docking programs coupled with suitable scoring functions are now established and very useful tools enabling computational chemists to rapidly screen large chem. databases and thereby to identify promising candidate compds. for further exptl. processing. In a broader scenario, predicting binding affinity is one of the most crit. and challenging components of computer-aided structure-based drug design. The development of a mol. docking scoring function which in principle could combine both features, namely ranking putative poses and predicting complex affinity, would be of paramount importance. Here, we systematically investigated the performance of the MM-PBSA approach, using two different Poisson-Boltzmann solvers (APBS and DelPhi), in the currently rising field of protein-peptide interactions (PPIs), identifying the correct binding conformations of 19 different protein-peptide complexes and predicting their binding free energies. First, we scored the decoy structures from HADDOCK calcn. via the MM-PBSA approach in order to assess the capability of retrieving near-native poses in the best-scoring clusters and of evaluating the corresponding free energies of binding. MM-PBSA behaves well in finding the poses corresponding to the lowest binding free energy. In order to improve the MM-PBSA-based scoring function, we dampened the MM-PBSA solvation and coulombic terms by 0.2, as proposed in the HADDOCK score and LIE approaches.
- 62Li, W.; Zhang, C.; Sui, J.; Kuhn, J. H.; Moore, M. J.; Luo, S.; Wong, S. K.; Huang, I. C.; Xu, K.; Vasilieva, N.; Murakami, A.; He, Y.; Marasco, W. A.; Guan, Y.; Choe, H.; Farzan, M. Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2. EMBO J. 2005, 24, 1634– 1643, DOI: 10.1038/sj.emboj.7600640100https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXktFequ7o%253D&md5=7b7d477b5376c04a7607f507b63702f2Receptor and viral determinants of SARS-coronavirus adaptation to human ACE2Li, Wenhui; Zhang, Chengsheng; Sui, Jianhua; Kuhn, Jens H.; Moore, Michael J.; Luo, Shiwen; Wong, Swee-Kee; Huang, I-Chueh; Xu, Keming; Vasilieva, Natalya; Murakami, Akikazu; He, Yaqing; Marasco, Wayne A.; Guan, Yi; Choe, Hyeryun; Farzan, MichaelEMBO Journal (2005), 24 (8), 1634-1643CODEN: EMJODG; ISSN:0261-4189. (Nature Publishing Group)Human angiotensin-converting enzyme 2 (ACE2) is a functional receptor for SARS coronavirus (SARS-CoV). Here we identify the SARS-CoV spike (S)-protein-binding site on ACE2. We also compare S proteins of SARS-CoV isolated during the 2002-2003 SARS outbreak and during the much less severe 2003-2004 outbreak, and from palm civets, a possible source of SARS-CoV found in humans. All three S proteins bound to and utilized palm-civet ACE2 efficiently, but the latter two S proteins utilized human ACE2 markedly less efficiently than did the S protein obtained during the earlier human outbreak. The lower affinity of these S proteins could be complemented by altering specific residues within the S-protein-binding site of human ACE2 to those of civet ACE2, or by altering S-protein residues 479 and 487 to residues conserved during the 2002-2003 outbreak. Collectively, these data describe mol. interactions important to the adaptation of SARS-CoV to human cells, and provide insight into the severity of the 2002-2003 SARS epidemic.
- 63Elbe, S.; Buckland-Merrett, G. Data, disease and diplomacy: GISAID’s innovative contribution to global health. Global Challenges 2017, 1, 33, DOI: 10.1002/gch2.101862https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MnjtFKmug%253D%253D&md5=ec5a87c7612a5922dd7bd76718359f20Data, disease and diplomacy: GISAID's innovative contribution to global healthElbe Stefan; Buckland-Merrett GemmaGlobal challenges (Hoboken, NJ) (2017), 1 (1), 33-46 ISSN:.The international sharing of virus data is critical for protecting populations against lethal infectious disease outbreaks. Scientists must rapidly share information to assess the nature of the threat and develop new medical countermeasures. Governments need the data to trace the extent of the outbreak, initiate public health responses, and coordinate access to medicines and vaccines. Recent outbreaks suggest, however, that the sharing of such data cannot be taken for granted - making the timely international exchange of virus data a vital global challenge. This article undertakes the first analysis of the Global Initiative on Sharing All Influenza Data as an innovative policy effort to promote the international sharing of genetic and associated influenza virus data. Based on more than 20 semi-structured interviews conducted with key informants in the international community, coupled with analysis of a wide range of primary and secondary sources, the article finds that the Global Initiative on Sharing All Influenza Data contributes to global health in at least five ways: (1) collating the most complete repository of high-quality influenza data in the world; (2) facilitating the rapid sharing of potentially pandemic virus information during recent outbreaks; (3) supporting the World Health Organization's biannual seasonal flu vaccine strain selection process; (4) developing informal mechanisms for conflict resolution around the sharing of virus data; and (5) building greater trust with several countries key to global pandemic preparedness.
- 64Baskaran, K.; Duarte, J. M.; Biyani, N.; Bliven, S.; Capitani, G. A PDB-wide, evolution-based assessment of protein-protein interfaces. BMC Struct. Biol. 2014, 14, 1422, DOI: 10.1186/s12900-014-0022-0There is no corresponding record for this reference.
- 65Amaral, M.; Kokh, D. B.; Bomke, J.; Wegener, A.; Buchstaller, H. P.; Eggenweiler, H. M. Protein conformational flexibility modulates kinetics and thermodynamics of drug binding. Nat. Commun. 2017, 8, 2276, DOI: 10.1038/s41467-017-02258-w64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mzksleqsg%253D%253D&md5=2ccf76a6569363af27b6f27cb1be5b7aProtein conformational flexibility modulates kinetics and thermodynamics of drug bindingAmaral M; Matias P; Amaral M; Wegener A; Frech M; Amaral M; Kokh D B; Wade R C; Bomke J; Buchstaller H P; Eggenweiler H M; Matias P; Sirrenberg C; Wade R C; Wade R CNature communications (2017), 8 (1), 2276 ISSN:.Structure-based drug design has often been restricted by the rather static picture of protein-ligand complexes presented by crystal structures, despite the widely accepted importance of protein flexibility in biomolecular recognition. Here we report a detailed experimental and computational study of the drug target, human heat shock protein 90, to explore the contribution of protein dynamics to the binding thermodynamics and kinetics of drug-like compounds. We observe that their binding properties depend on whether the protein has a loop or a helical conformation in the binding site of the ligand-bound state. Compounds bound to the helical conformation display slow association and dissociation rates, high-affinity and high cellular efficacy, and predominantly entropically driven binding. An important entropic contribution comes from the greater flexibility of the helical relative to the loop conformation in the ligand-bound state. This unusual mechanism suggests increasing target flexibility in the bound state by ligand design as a new strategy for drug discovery.
- 66Ferina, J.; Daggett, V. Visualizing protein folding and unfolding. J. Mol. Biol. 2019, 431, 1540– 1564, DOI: 10.1016/j.jmb.2019.02.02665https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXksFKnuro%253D&md5=4296169975d3fdd59d279bb7afd26303Visualizing Protein Folding and UnfoldingFerina, Jennifer; Daggett, ValerieJournal of Molecular Biology (2019), 431 (8), 1540-1564CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A review. Protein folding/unfolding is a complicated process that defies high-resoln. characterization by exptl. methods. As an alternative, atomistic mol. dynamics simulations are now routinely employed to elucidate and magnify the accompanying conformational changes and the role of solvent in the folding process. However, the level of detail necessary to map the process at high spatial-temporal resoln. provides an overwhelming amt. of data. As more and better tools are developed for anal. of these large data sets and validation of the simulations, one is still left with the problem of visualizing the results in ways that provide insight into the folding/unfolding process. While viewing and interrogating static crystal structures has become commonplace, more and different approaches are required for dynamic, interconverting, unfolding, and refolding proteins. Here the authors review a variety of approaches, ranging from straightforward to complex and unintuitive for multiscale anal. and visualization of protein folding and unfolding.
- 67Zambrano, R.; Jamroz, M.; Szczasiuk, A.; Pujols, J.; Kmiecik, S.; Ventura, S. AGGRESCAN3D (A3D): server for prediction of aggregation properties of protein structures. Nucleic Acids Res. 2015, 43, W306– 313, DOI: 10.1093/nar/gkv35966https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVymtbjK&md5=4d5a4d94fa0bf2744250860780e2a203AGGRESCAN3D (A3D): server for prediction of aggregation properties of protein structuresZambrano, Rafael; Jamroz, Michal; Szczasiuk, Agata; Pujols, Jordi; Kmiecik, Sebastian; Ventura, SalvadorNucleic Acids Research (2015), 43 (W1), W306-W313CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Protein aggregation underlies an increasing no. of disorders and constitutes a major bottleneck in the development of therapeutic proteins. Our present understanding on the mol. determinants of protein aggregation has crystd. in a series of predictive algorithms to identify aggregation-prone sites. A majority of these methods rely only on sequence. Therefore, they find difficulties to predict the aggregation properties of folded globular proteins, where aggregation-prone sites are often not contiguous in sequence or buried inside the native structure. The AGGRESCAN3D (A3D) server overcomes these limitations by taking into account the protein structure and the exptl. aggregation propensity scale from the well-established AGGRESCAN method. Using the A3D server, the identified aggregation-prone residues can be virtually mutated to design variants with increased soly., or to test the impact of pathogenic mutations. Addnl., A3D server enables to take into account the dynamic fluctuations of protein structure in soln., which may influence aggregation propensity. This is possible in A3D Dynamic Mode that exploits the CABS-flex approach for the fast simulations of flexibility of globular proteins.
- 68Goel, B.; Bhardwaj, N.; Tripathi, N.; Jain, S. K. Drug discovery of small molecules for the treatment of COVID-19: a review on clinical studies. Mini-Rev. Med. Chem. 2021, 21, 1431, DOI: 10.2174/138955752166620122814575567https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1Sjt7vP&md5=05d78cb3bc1278ab6ec389f4c403a0f8Drug Discovery of Small Molecules for the Treatment of COVID-19: A Review on Clinical StudiesGoel, Bharat; Bhardwaj, Nivedita; Tripathi, Nancy; Jain, Shreyans K.Mini-Reviews in Medicinal Chemistry (2021), 21 (12), 1431-1456CODEN: MMCIAE; ISSN:1389-5575. (Bentham Science Publishers Ltd.)A review. Recently, a sudden outbreak of novel coronavirus disease (COVID-19) was caused by a zoonotic virus known as severe acute respiratory syndrome coronavirus (SARS-CoV-2). It has caused pandemic situations around the globe affecting the lives of millions of people. So far, no drug has been approved for the treatment of SARS-CoV-2 infected patients. As of now, more than 1000 clin. trials are going on for repurposing of FDA-approved drugs and for evaluating the safety and efficiency of exptl. antiviral mols. to combat COVID-19. Since the development of new drugs may require months to years to reach the market, this review focusses on the potential of existing small mol. FDA approved drugs and the mols. already in the clin. pipeline against viral infections like HIV, hepatitis B, Ebola virus, and other viruses of coronavirus family (SARS-CoV and MERS-CoV). The review also discusses the natural products and traditional medicines in clin. studies against COVID-19. Currently, 1978 studies are active, 143 completed and 4 posted results (as of June 13, 2020) on clinicaltrials.gov.
- 69Pant, S.; Singh, M.; Ravichandiran, V.; Murty, U. S. N.; Srivastava, H. K. Peptide-like and small-molecule inhibitors against COVID-19. J. Biomol. Struct. Dyn. 2021, 39, 2904, DOI: 10.1080/07391102.2020.175751068https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVShurrO&md5=91f4cbaeddbc425f807f2ee413023d2cPeptide-like and small-molecule inhibitors against Covid-19Pant, Suyash; Singh, Meenakshi; Ravichandiran, V.; Murty, U. S. N.; Srivastava, Hemant KumarJournal of Biomolecular Structure and Dynamics (2021), 39 (8), 2904-2913CODEN: JBSDD6; ISSN:0739-1102. (Taylor & Francis Ltd.)Coronavirus disease strain (SARS-CoV-2) was discovered in 2019, and it is spreading very fast around the world causing the disease Covid-19. Currently, >1.6 million individuals are infected, and several thousand are dead across the globe because of Covid-19. We utilized the in-silico approaches to identify possible protease inhibitors against SARS-CoV-2. Potential compds. were screened from the CHEMBL database, ZINC database, FDA approved drugs, and mols. under clin. trials. Our study is based on 6Y2F and 6W63 co-crystd. structures available in the protein data bank (PDB). Seven hundred compds. from ZINC/CHEMBL databases and 1400 compds. from drug-bank were selected based on pos. interactions with the reported binding site. All the selected compds. were subjected to std.-precision (SP) and extra-precision (XP) mode of docking. Generated docked poses were carefully visualized for known interactions within the binding site. Mol. mechanics-generalized born surface area (MM-GBSA) calcns. were performed to screen the best compds. based on docking scores and binding energy values. Mol. dynamics (MD) simulations were carried out on 4 selected compds. from the CHEMBL database to validate the stability and interactions. MD simulations were also performed on the PDB structure 6YF2F to understand the differences between screened mols. and co-crystd. ligand. We screened 300 potential compds. from various databases, and 66 potential compds. from FDA approved drugs. Cobicistat, ritonavir, lopinavir, and darunavir are in the top screened mols. from FDA approved drugs. The screened drugs and mols. may be helpful in fighting with SARS-CoV-2 after further studies.
- 70Di Natale, C.; La Manna, S.; De Benedictis, I.; Brandi, P.; Marasco, D. Perspectives in peptide-based vaccination strategies for syndrome coronavirus 2 pandemic. Front. Pharmacol. 2020, 11, 578382, DOI: 10.3389/fphar.2020.57838269https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjsl2jsLg%253D&md5=c1b4fea4454c0207169e74a870617bbfPerspectives in peptide-based vaccination strategies for syndrome coronavirus 2 pandemicDi Natale, Concetta; La Manna, Sara; De Benedictis, Ilaria; Brandi, Paola; Marasco, DanielaFrontiers in Pharmacology (2020), 11 (), 578382CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)At the end of Dec. 2019, an epidemic form of respiratory tract infection now named COVID-19 emerged in Wuhan, China. It is caused by a newly identified viral pathogen, the severe acute respiratory syndrome coronavirus (SARS-CoV-2), which can cause severe pneumonia and acute respiratory distress syndrome. On Jan. 30, 2020, due to the rapid spread of infection, COVID-19 was declared as a global health emergency by the World Health Organization. Coronaviruses are enveloped RNA viruses belonging to the family of Coronaviridae, which are able to infect birds, humans and other mammals. The majority of human coronavirus infections are mild although already in 2003 and in 2012, the epidemics of SARS-CoV and Middle East Respiratory Syndrome coronavirus (MERSCoV), resp., were characterized by a high mortality rate. In this regard, many efforts have been made to develop therapeutic strategies against human CoV infections but, unfortunately, drug candidates have shown efficacy only into in vitro studies, limiting their use against COVID-19 infection. Actually, no treatment has been approved in humans against SARS-CoV-2, and therefore there is an urgent need of a suitable vaccine to tackle this health issue. However, the puzzled scenario of biol. features of the virus and its interaction with human immune response, represent a challenge for vaccine development. As expected, in hundreds of research labs. there is a running out of breath to explore different strategies to obtain a safe and quickly spreadable vaccine; and among others, the peptide-based approach represents a turning point as peptides have demonstrated unique features of selectivity and specificity toward specific targets. Peptide-based vaccines imply the identification of different epitopes both on human cells and virus capsid and the design of peptide/peptidomimetics able to counteract the primary host-pathogen interaction, in order to induce a specific host immune response. SARS-CoV-2 immunogenic regions are mainly distributed, as well as for other coronaviruses, across structural areas such as spike, envelope, membrane or nucleocapsid proteins. Herein, we aim to highlight the mol. basis of the infection and recent peptide-based vaccines strategies to fight the COVID-19 pandemic including their delivery systems.
- 71Pessi, A.; Bixler, S. L.; Soloveva, V.; Radoshitzky, S.; Retterer, C. Cholesterol-conjugated stapled peptides inhibit Ebola and Marburg viruses in vitro and in vivo. Antiviral Res. 2019, 171, 104592, DOI: 10.1016/j.antiviral.2019.10459270https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVSgs7fO&md5=a5ca9bbc605a0e0618473ce5e7a3728fCholesterol-conjugated stapled peptides inhibit Ebola and Marburg viruses in vitro and in vivoPessi, Antonello; Bixler, Sandra L.; Soloveva, Veronica; Radoshitzky, Sheli; Retterer, Cary; Kenny, Tara; Zamani, Rouzbeh; Gomba, Glenn; Gharabeih, Dima; Wells, Jay; Wetzel, Kelly S.; Warren, Travis K.; Donnelly, Ginger; Van Tongeren, Sean A.; Steffens, Jesse; Duplantier, Allen J.; Kane, Christopher D.; Vicat, Pascale; Couturier, Valerie; Kester, Kent E.; Shiver, John; Carter, Kara; Bavari, SinaAntiviral Research (2019), 171 (), 104592CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)Currently, there are no therapeutics approved and the need for Ebola-specific therapeutics remains a gap. In search for anti-Ebola therapies we tested the idea of using inhibitory properties of peptides corresponding to the C-terminal heptad-repeat (HR2) domains of class I fusion proteins against EBOV infection. The fusion protein GP2 of EBOV belongs to class I, suggesting that a similar strategy to HIV may be applied to inhibit EBOV infection. The serum half-life of peptides was expanded by cholesterol conjugation to allow daily dosing. The peptides were further constrained to stabilize a helical structure to increase the potency of inhibition. The EC50s of lead peptides were in low micromolar range, as detd. by a high-content imaging test of EBOV-infected cells. Lead peptides were tested in an EBOV lethal mouse model and efficacy of the peptides were detd. following twice-daily administration of peptides for 9 days. The most potent peptide was able to protect mice from lethal challenge of mouse-adapted Ebola virus. These data show that engineered peptides coupled with cholesterol can inhibit viral prodn., protect mice against lethal EBOV infection, and may be used to build novel therapeutics against EBOV.
- 72Li, C. G.; Tang, W.; Chi, X. J.; Dong, Z. M.; Wang, X. X.; Wang, X. J. A cholesterol tag at the N terminus of the relatively broad-spectrum fusion inhibitory peptide targets an earlier stage of fusion glycoprotein activation and increases the peptide’s antiviral potency in vivo. J. Virol. 2013, 87, 9223– 9232, DOI: 10.1128/JVI.01153-1371https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1WgtLvP&md5=d418cb40ba11ea15d7c50ad61ee2caaaA cholesterol tag at the N terminus of the relatively broad-spectrum fusion inhibitory peptide targets an earlier stage of fusion glycoprotein activation and increases the peptide's antiviral potency in vivoLi, Chuan-Gen; Wang, Tang; Chi, Xiao-Jing; Dong, Zhi-Ming; Wang, Xi-Xi; Wang, Xiao-JiaJournal of Virology (2013), 87 (16), 9223-9232CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)In previous work, we designed peptides that showed potent inhibition of Newcastle disease virus (NDV) and infectious bronchitis virus (IBV) infections in chicken embryos. In this study, we demonstrate that peptides modified with cholesterol or 3 U of polyethylene glycol (PEG3) conjugated to the peptides' N termini showed even more promising antiviral activities when tested in animal models. Both cholesterol- and cholesterol-PEG3-tagged peptides were able to protect chicken embryos from infection with different serotypes of NDV and IBV when administered 12 h prior to virus inoculation. In comparison, the untagged peptides required intervention closer to the time of viral inoculation to achieve a similar level of protection. I.m. injection of cholesterol-tagged peptide at 1.6 mg/kg 1 day before virus infection and then three times at 3-day intervals after viral inoculation protected 70% of the chickens from NDV infection. We further demonstrate that the cholesterol-tagged peptide has an in vivo half-life greater than that of untagged peptides. It also has the potential to cross the blood-brain barrier to enter the avian central nervous system (CNS). Finally, we show that the cholesterol-tagged peptide could play a role before the viral fusion peptide's insertion into the host cell and thereby target an earlier stage of fusion glycoprotein activation. Our findings are of importance for the further development of antivirals with broad-spectrum protective effects.
- 73Pessi, A. Cholesterol-conjugated peptide antivirals: a path to a rapid response to emerging viral diseases. J. Pept. Sci. 2015, 21, 379– 386, DOI: 10.1002/psc.270672https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntFCkt7w%253D&md5=56d038b929227c81bdc5e85cc562de75Cholesterol-conjugated peptide antivirals: a path to a rapid response to emerging viral diseasesPessi, AntonelloJournal of Peptide Science (2015), 21 (5), 379-386CODEN: JPSIEI; ISSN:1075-2617. (John Wiley & Sons Ltd.)A review. While it is now possible to identify and genetically fingerprint the causative agents of emerging viral diseases, often with extraordinary speed, suitable therapies cannot be developed with equiv. speed, because drug discovery requires information that goes beyond knowledge of the viral genome. Peptides, however, may represent a special opportunity. For all enveloped viruses, fusion between the viral and the target cell membrane is an obligatory step of the life cycle. Class I fusion proteins harbor regions with a repeating pattern of amino acids, the heptad repeats (HRs), that play a key role in fusion, and HR-derived peptides such as enfuvirtide, in clin. use for HIV, can block the process. Because of their characteristic sequence pattern, HRs are easily identified in the genome by means of computer programs, providing the sequence of candidate peptide inhibitors directly from genomic information. Moreover, a simple chem. modification, the attachment of a cholesterol group, can dramatically increase the antiviral potency of HR-derived inhibitors and simultaneously improve their pharmacokinetics. Further enhancement can be provided by dimerization of the cholesterol-conjugated peptide. The examples reported so far include inhibitors of retroviruses, paramyxoviruses, orthomyxoviruses, henipaviruses, coronaviruses, and filoviruses. For some of these viruses, in vivo efficacy has been demonstrated in suitable animal models. The combination of bioinformatic lead identification and potency/pharmacokinetics improvement provided by cholesterol conjugation may form the basis for a rapid response strategy, where development of an emergency cholesterol-conjugated therapeutic would immediately follow the availability of the genetic information of a new enveloped virus. Copyright © 2014 European Peptide Society and John Wiley & Sons, Ltd.
- 74Pettersen, E. F.; Goddard, T. D.; Huang, C. C.; Couch, G. S.; Greenblatt, D. M.; Meng, E. C.; Ferrin, T. E. UCSF chimera--a visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605– 1612, DOI: 10.1002/jcc.2008473https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmvVOhsbs%253D&md5=944b175f440c1ff323705987cf937ee7UCSF Chimera-A visualization system for exploratory research and analysisPettersen, Eric F.; Goddard, Thomas D.; Huang, Conrad C.; Couch, Gregory S.; Greenblatt, Daniel M.; Meng, Elaine C.; Ferrin, Thomas E.Journal of Computational Chemistry (2004), 25 (13), 1605-1612CODEN: JCCHDD; ISSN:0192-8651. (John Wiley & Sons, Inc.)The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale mol. assemblies such as viral coats, and Collab., which allows researchers to share a Chimera session interactively despite being at sep. locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and assocd. structures; ViewDock, for screening docked ligand orientations; Movie, for replaying mol. dynamics trajectories; and Vol. Viewer, for display and anal. of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/.
- 75Caporale, A.; Doti, N.; Monti, A.; Sandomenico, A.; Ruvo, M. Automatic procedures for the synthesis of difficult peptides using oxyma as activating reagent: A comparative study on the use of bases and on different deprotection and agitation conditions. Peptides 2018, 102, 38– 46, DOI: 10.1016/j.peptides.2018.02.00674https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjvVClsbw%253D&md5=e62fa59f680ab328407d6992db13b802Automatic procedures for the synthesis of difficult peptides using oxyma as activating reagent: A comparative study on the use of bases and on different deprotection and agitation conditionsCaporale, A.; Doti, N.; Monti, A.; Sandomenico, A.; Ruvo, M.Peptides (New York, NY, United States) (2018), 102 (), 38-46CODEN: PPTDD5; ISSN:0196-9781. (Elsevier)Solid-Phase Peptide Synthesis (SPPS) is a rapid and efficient methodol. for the chem. synthesis of peptides and small proteins. However, the assembly of peptide sequences classified as "difficult" poses severe synthetic problems in SPPS for the occurrence of extensive aggregation of growing peptide chains which often leads to synthesis failure. In this framework, we have investigated the impact of different synthetic procedures on the yield and final purity of three well-known "difficult peptides" prepd. using oxyma as additive for the coupling steps. In particular, we have comparatively investigated the use of piperidine and morpholine/DBU as deprotection reagents, the addn. of DIPEA, collidine and N-methylmorpholine as bases to the coupling reagent. Moreover, the effect of different agitation modalities during the acylation reactions has been investigated. Data obtained represent a step forward in optimizing strategies for the synthesis of "difficult peptides".
- 76Grimsley, G. R.; Pace, C. N. Spectrophotometric Determination of Protein Concentration. Curr. Protoc. Protein Sci. 2003, 3.1.1– 3.1.9, DOI: 10.1002/0471140864.ps0301s33There is no corresponding record for this reference.
- 77Scopes, R.K. Measurement of protein by spectrophotometry at 205 nm. Anal. Biochem. 1974, 59, 277– 282, DOI: 10.1016/0003-2697(74)90034-776https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE2cXksVOjsb0%253D&md5=ff53a6ad935bde82fc8c8a60bb606760Measurement of protein by spectrophotometry at 205nmScopes, R. K.Analytical Biochemistry (1974), 59 (1), 277-82CODEN: ANBCA2; ISSN:0003-2697.A method was described for the measurement of protein concn. by using the peptide bond absorption at 205 nm. The extinction coeff. (ε) at 205 nm was estd., allowing for the absorption due to tryptophan and tyrosine residues, by measuring the absorbance at 280 nm as well as at 205 nm. The estd. ε205 was compared with the actual ε205 for a no. of proteins, the mean error being <2%. This was about 3 times better than using an av. ε2051mg/ml of 31 and approached the range of exptl. error inherent in any method of protein estn.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.1c00477.
Additional figures illustrating molecular interaction from MD analysis of designed peptides and S-RBD, binding free energy of ACE2 derived peptides in complexes of S-RBD (binding energies of ACE2:S-RBD included for comparison), peptide LC-MS characterization, details of changes in hot spot area plot caused by aggregation of peptides, variation of fluorescence peak intensity with increasing concentration of peptides, CD spectra of peptides, overlay of emission spectra of samples dissolved in PBS and PBST, 1% (w/v) PEG8000 at 25 μM in 2.5% DMSO, and SEC of peptides; additional tables detailing key interactions between S-RBD and ACE2 identified using PiPreD, peptide sequences including replacement of some residues for improved targeting of S-RBD, analysis of data collected for the calculation of concentration of peptides in PBS and PBST, 1% (w/v) PEG8000 by UV spectroscopy analysis, theoretical, apparent molecular weight, and Rt values of ACE2-antagonist peptides as determined by SEC analysis, and dose–response results of the antiviral activity of peptides in a SARS-CoV-2 infection inhibition assay performed in VERO-E6 cells (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.