1
|
Islam MA, Pathak K, Saikia R, Pramanik P, Das A, Talukdar P, Shakya A, Ghosh SK, Singh UP, Bhat HR. An in-depth analysis of COVID-19 treatment: Present situation and prospects. Arch Pharm (Weinheim) 2024; 357:e2400307. [PMID: 39106224 DOI: 10.1002/ardp.202400307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 08/09/2024]
Abstract
Coronavirus disease 2019 (COVID-19) the most contagious infection caused by the unique type of coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), produced a global pandemic that wreaked havoc on the health-care system, resulting in high morbidity and mortality. Several methods were implemented to tackle the virus, including the repurposing of existing medications and the development of vaccinations. The purpose of this article is to provide a complete summary of the current state and future possibilities for COVID-19 therapies. We describe the many treatment classes, such as antivirals, immunomodulators, and monoclonal antibodies, that have been repurposed or developed to treat COVID-19. We also looked at the clinical evidence for these treatments, including findings from observational studies and randomized-controlled clinical trials, and highlighted the problems and limitations of the available evidence. Furthermore, we reviewed existing clinical trials and prospective COVID-19 therapeutic options, such as novel medication candidates and combination therapies. Finally, we discussed the long-term consequences of COVID-19 and the importance of ongoing research into the development of viable treatments. This review will help physicians, researchers, and policymakers to understand the prevention and mitigation of COVID-19.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Kalyani Pathak
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Riya Saikia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Pallab Pramanik
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Aparoop Das
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Prasenjit Talukdar
- Department of Petroleum Engineering, DUIET, Dibrugarh, University, Assam, India
| | - Anshul Shakya
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Surajit Kumar Ghosh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Udaya Pratap Singh
- Drug Design & Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh, India
| | - Hans Raj Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| |
Collapse
|
2
|
Osman AMA, Arabi AA. Average Electron Density: A Quantitative Tool for Evaluating Non-Classical Bioisosteres of Amides. ACS OMEGA 2024; 9:13172-13182. [PMID: 38524460 PMCID: PMC10955596 DOI: 10.1021/acsomega.3c09732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 03/26/2024]
Abstract
Bioisosterism is strategically used in drug design to enhance the pharmacokinetic and pharmacodynamic properties of therapeutic molecules. The average electron density (AED) tool has been used in several studies to quantify similarities among nonclassical bioisosteres of carboxylic acid. In this study, the AED tool is used to quantify the similarities among nonclassical bioisosteres of an amide group. In particular, amide-to-1,2,3-triazole bioisosterism is considered. To evaluate the AED differences exhibited by isomers of nonclassical bioisosteres, both isomers of amide (cis and trans) and 1,2,3-triazole (1,4 and 1,5 disubstituted moieity) were considered. The amide and 1,2,3-triazole bioisosteric moieties were capped with various R groups (R= methyl, hydrogen, and chloro) to account for changes in their environment. Amide-to-triazole bioisosteric substitutions were then explored in a more realistic environment, that is, within the FDA-approved anticancer imatinib drug. The AED tool effectively identified similarities between substantially different moieties, 1,2,3-triazole and amide, showing AED differences of no more than 4%. The AED tool was also proven to be useful in evaluating the contribution of various factors affecting triazole-amide bioisosterism including isomerism and changes in their environment. The AED values of each bioisostere were transferable within a maximum difference of 2.6%, irrespective of the change in environment. The 1,4- and 1,5-disubstituted isomers of 1,2,3-triazole have AED values that differ by less than unity, 0.52%. Similarly, the AED values of the cis- and trans-amide isomers differ by only 1.31%. Overall, the AED quantitative tool not only replicated experimental observations regarding similarities in bioisosteres, but also explained and quantified each contributing factor. This demonstrates the extended utility of the AED tool from nonclassical carboxylic acid bioisosteres to amide equivalents.On the contrary, electrostatic potential maps, usually used in the literature to qualitatively evaluate bioisosterism, were not similar for the 1,2,3-triazole and amide bioisosteres, under different environments. Overall, the AED tool proves to be powerful in quantitatively evaluating and predicting bioisosterism across diverse moieties considering structural and environmental variations, making it valuable in drug design.
Collapse
Affiliation(s)
- Alaa MA Osman
- College of Medicine and Health
Sciences, Department of Biochemistry and Molecular Biology, United Arab Emirates University, AlAin P.O. Box: 15551, United Arab Emirates
| | - Alya A. Arabi
- College of Medicine and Health
Sciences, Department of Biochemistry and Molecular Biology, United Arab Emirates University, AlAin P.O. Box: 15551, United Arab Emirates
| |
Collapse
|
3
|
Bhole RP, Patil S, Kapare HS, Chikhale RV, Gurav SS. PROTAC Beyond Cancer- Exploring the New Therapeutic Potential of Proteolysis Targeting Chimeras. Curr Top Med Chem 2024; 24:2050-2073. [PMID: 38963108 DOI: 10.2174/0115680266309968240621072550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024]
Abstract
In the realm of oncology, the transformative impact of PROTAC (PROteolysis TAgeting Chimeras) technology has been particularly pronounced since its introduction in the 21st century. Initially conceived for cancer treatment, PROTACs have evolved beyond their primary scope, attracting increasing interest in addressing a diverse array of medical conditions. This expanded focus includes not only oncological disorders but also viral infections, bacterial ailments, immune dysregulation, neurodegenerative conditions, and metabolic disorders. This comprehensive review explores the broadening landscape of PROTAC application, highlighting ongoing developments and innovations aimed at deploying these molecules across a spectrum of diseases. Careful consideration of the design challenges associated with PROTACs reveals that, when appropriately addressed, these compounds present significant advantages over traditional therapeutic approaches, positioning them as promising alternatives. To evaluate the efficacy of PROTAC molecules, a diverse array of assays is employed, ranging from High-Throughput Imaging (HTI) assays to Cell Painting assays, CRBN engagement assays, Fluorescence Polarization assays, amplified luminescent proximity homogeneous assays, Timeresolved fluorescence energy transfer assays, and Isothermal Titration Calorimetry assays. These assessments collectively contribute to a nuanced understanding of PROTAC performance. Looking ahead, the trajectory of PROTAC technology suggests its potential recognition as a versatile therapeutic strategy for an expansive range of medical conditions. Ongoing progress in this field sets the stage for PROTACs to emerge as valuable tools in the multifaceted landscape of medical treatments.
Collapse
Affiliation(s)
- Ritesh P Bhole
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, India
- Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyappeth, Pimpri, Pune, 411018, India
| | - Sapana Patil
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, India
| | - Harshad S Kapare
- Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018, India
| | | | - Shailendra S Gurav
- Department of Pharmacognosy, Goa College of Pharmacy, Panjim, Goa, India
| |
Collapse
|
4
|
Girgis AS, Panda SS, Kariuki BM, Bekheit MS, Barghash RF, Aboshouk DR. Indole-Based Compounds as Potential Drug Candidates for SARS-CoV-2. Molecules 2023; 28:6603. [PMID: 37764378 PMCID: PMC10537473 DOI: 10.3390/molecules28186603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
The COVID-19 pandemic has posed a significant threat to society in recent times, endangering human health, life, and economic well-being. The disease quickly spreads due to the highly infectious SARS-CoV-2 virus, which has undergone numerous mutations. Despite intense research efforts by the scientific community since its emergence in 2019, no effective therapeutics have been discovered yet. While some repurposed drugs have been used to control the global outbreak and save lives, none have proven universally effective, particularly for severely infected patients. Although the spread of the disease is generally under control, anti-SARS-CoV-2 agents are still needed to combat current and future infections. This study reviews some of the most promising repurposed drugs containing indolyl heterocycle, which is an essential scaffold of many alkaloids with diverse bio-properties in various biological fields. The study also discusses natural and synthetic indole-containing compounds with anti-SARS-CoV-2 properties and computer-aided drug design (in silico studies) for optimizing anti-SARS-CoV-2 hits/leads.
Collapse
Affiliation(s)
- Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (R.F.B.); (D.R.A.)
| | - Siva S. Panda
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
| | - Benson M. Kariuki
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, UK; (B.M.K.)
| | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (R.F.B.); (D.R.A.)
| | - Reham F. Barghash
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (R.F.B.); (D.R.A.)
| | - Dalia R. Aboshouk
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (R.F.B.); (D.R.A.)
| |
Collapse
|
5
|
Liang J, Wu Y, Lan K, Dong C, Wu S, Li S, Zhou HB. Antiviral PROTACs: Opportunity borne with challenge. CELL INSIGHT 2023; 2:100092. [PMID: 37398636 PMCID: PMC10308200 DOI: 10.1016/j.cellin.2023.100092] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 07/04/2023]
Abstract
Proteolysis targeting chimera (PROTAC) degradation of pathogenic proteins by hijacking of the ubiquitin-proteasome-system has become a promising strategy in drug design. The overwhelming advantages of PROTAC technology have ensured a rapid and wide usage, and multiple PROTACs have entered clinical trials. Several antiviral PROTACs have been developed with promising bioactivities against various pathogenic viruses. However, the number of reported antiviral PROTACs is far less than that of other diseases, e.g., cancers, immune disorders, and neurodegenerative diseases, possibly because of the common deficiencies of PROTAC technology (e.g., limited available ligands and poor membrane permeability) plus the complex mechanism involved and the high tendency of viral mutation during transmission and replication, which may challenge the successful development of effective antiviral PROTACs. This review highlights the important advances in this rapidly growing field and critical limitations encountered in developing antiviral PROTACs by analyzing the current status and representative examples of antiviral PROTACs and other PROTAC-like antiviral agents. We also summarize and analyze the general principles and strategies for antiviral PROTAC design and optimization with the intent of indicating the potential strategic directions for future progress.
Collapse
Affiliation(s)
- Jinsen Liang
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Yihe Wu
- Provincial Key Laboratory of Developmentally Originated Disease, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Chune Dong
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Shuwen Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Shu Li
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Hai-Bing Zhou
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Provincial Key Laboratory of Developmentally Originated Disease, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
6
|
Nogara PA, Omage FB, Bolzan GR, Delgado CP, Orian L, Rocha JBT. Reactivity and binding mode of disulfiram, its metabolites, and derivatives in SARS-CoV-2 PL pro: insights from computational chemistry studies. J Mol Model 2022; 28:354. [PMID: 36222962 PMCID: PMC9554863 DOI: 10.1007/s00894-022-05341-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/28/2022] [Indexed: 10/25/2022]
Abstract
The papain-like protease (PLpro) from SARS-CoV-2 is an important target for the development of antivirals against COVID-19. The safe drug disulfiram (DSF) presents antiviral activity inhibiting PLpro in vitro, and it is under clinical trial studies, indicating to be a promising anti-COVID-19 drug. In this work, we aimed to understand the mechanism of PLpro inhibition by DSF and verify if DSF metabolites and derivatives could be potential inhibitors too. Molecular docking, DFT, and ADMET techniques were applied. The carbamoylation of the active site cysteine residue by DSF metabolite (DETC-MeSO) is kinetically and thermodynamically favorable (ΔG‡ = 3.15 and ΔG = - 12.10 kcal mol-1, respectively). Our results strongly suggest that the sulfoxide metabolites from DSF are promising covalent inhibitors of PLpro and should be tested in in vitro and in vivo assays to confirm their antiviral action.
Collapse
Affiliation(s)
- Pablo Andrei Nogara
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, 97105-900, Brazil.
- Instituto Federal de Educação Ciência E Tecnologia Farroupilha (IFFar), Rua Fabio João Andolhe 1100, Santo Augusto, RS, 98590-000, Brazil.
| | - Folorunsho Bright Omage
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Gustavo Roni Bolzan
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Cássia Pereira Delgado
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, 97105-900, Brazil
| | - Laura Orian
- Dipartimento Di Scienze Chimiche, Università Degli Studi Di Padova, Via Marzolo 1, 35131, Padua, Italy
| | - João Batista Teixeira Rocha
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Av. Roraima 1000, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
7
|
Youssef MA, Panda SS, Aboshouk DR, Said MF, El Taweel A, GabAllah M, Fayad W, Soliman AF, Mostafa A, Fawzy NG, Girgis AS. Novel Curcumin Mimics: Design, Synthesis, Biological Properties and Computational Studies of Piperidone‐Piperazine Conjugates. ChemistrySelect 2022. [DOI: 10.1002/slct.202201406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- M. Adel Youssef
- Department of Chemistry Faculty of Science Helwan University Helwan Egypt
| | - Siva S. Panda
- Department of Chemistry and Physics Augusta University Augusta GA 30912 USA
| | - Dalia R. Aboshouk
- Department of Pesticide Chemistry National Research Centre Dokki Giza 12622 Egypt
| | - Mona F. Said
- Department of Pharmaceutical Chemistry Faculty of Pharmacy Cairo University Cairo 11562 Egypt
| | - Ahmed El Taweel
- Center of Scientific Excellence for Influenza Viruses National Research Centre Dokki Giza 12622 Egypt
| | - Mohamed GabAllah
- Center of Scientific Excellence for Influenza Viruses National Research Centre Dokki Giza 12622 Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department National Research Centre Dokki, Giza 12622 Egypt
| | - Ahmed F. Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department National Research Centre Dokki, Giza 12622 Egypt
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses National Research Centre Dokki Giza 12622 Egypt
| | - Nehmedo G. Fawzy
- Department of Pesticide Chemistry National Research Centre Dokki Giza 12622 Egypt
| | - Adel S. Girgis
- Department of Pesticide Chemistry National Research Centre Dokki Giza 12622 Egypt
| |
Collapse
|
8
|
Concise synthesis of antiviral drug, Molnupiravir by direct coupling of fully protected D-Ribose with cytosine. Tetrahedron Lett 2022. [DOI: 10.1016/j.tetlet.2022.153783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
9
|
Abstract
Identification of an effective antiviral for the treatment of COVID-19 is considered one of the holy grails in the bid to end the pandemic. However, the novelty of SARS-CoV-2, along with the little knowledge available about its infection characteristics at the beginning of this pandemic, challenges the scientific world on how one may be able to promptly identify promising drug candidates from a myriad of compound libraries. Here, we describe a cytopathic effect (CPE)-based drug screening assay for SARS-CoV-2 which allows for rapid assessment of drug compound libraries through pre- or posttreatment drug screening procedures and evaluation using a light microscope. By comparing the virus-induced CPE of the drug-treated cells against the vehicle and drug controls, potent drug candidates can be quickly identified for further downstream studies.
Collapse
Affiliation(s)
- Yan Ling Ng
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chee Keng Mok
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Collaborative and Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
10
|
Desantis J, Mercorelli B, Celegato M, Croci F, Bazzacco A, Baroni M, Siragusa L, Cruciani G, Loregian A, Goracci L. Indomethacin-based PROTACs as pan-coronavirus antiviral agents. Eur J Med Chem 2021; 226:113814. [PMID: 34534839 PMCID: PMC8416298 DOI: 10.1016/j.ejmech.2021.113814] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 11/29/2022]
Abstract
Indomethacin (INM), a well-known non-steroidal anti-inflammatory drug, has recently gained attention for its antiviral activity demonstrated in drug repurposing studies against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Although the mechanism of action of INM is not yet fully understood, recent studies have indicated that it acts at an early stage of the coronaviruses (CoVs) replication cycle. In addition, a proteomic study reported that the anti-SARS-CoV-2 activity of INM could be also ascribed to its ability to inhibit human prostaglandin E synthase type 2 (PGES-2), a host protein which interacts with the SARS-CoV-2 NSP7 protein. Although INM does not potently inhibit SARS-CoV-2 replication in infected Vero E6 cells, here we have explored for the first time the application of the Proteolysis Targeting Chimeras (PROTACs) technology in order to develop more potent INM-derived PROTACs with anti-CoV activity. In this study, we report the design, synthesis, and biological evaluation of a series of INM-based PROTACs endowed with antiviral activity against a panel of human CoVs, including different SARS-CoV-2 strains. Two PROTACs showed a strong improvement in antiviral potency compared to INM. Molecular modelling studies support human PGES-2 as a potential target of INM-based antiviral PROTACs, thus paving the way toward the development of host-directed anti-CoVs strategies. To the best of our knowledge, these PROTACs represent the first-in-class INM-based PROTACs with antiviral activity and also the first example of the application of PROTACs to develop pan-coronavirus agents.
Collapse
Affiliation(s)
- Jenny Desantis
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Marta Celegato
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Federico Croci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Italy
| | | | - Massimo Baroni
- Molecular Discovery Ltd., Centennial Park, Borehamwood, Hertfordshire, United Kingdom
| | | | - Gabriele Cruciani
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| | - Laura Goracci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Italy.
| |
Collapse
|
11
|
Srour AM, Panda SS, Mostafa A, Fayad W, El-Manawaty MA, A. F. Soliman A, Moatasim Y, El Taweel A, Abdelhameed MF, Bekheit MS, Ali MA, Girgis AS. Synthesis of aspirin-curcumin mimic conjugates of potential antitumor and anti-SARS-CoV-2 properties. Bioorg Chem 2021; 117:105466. [PMID: 34775204 PMCID: PMC8566089 DOI: 10.1016/j.bioorg.2021.105466] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/31/2021] [Indexed: 12/16/2022]
Abstract
Series of piperidone-salicylate conjugates were synthesized through the reaction of 3E,5E-bis(arylidene)-4-piperidones with the appropriate acid chloride of acetylsalicylate in the presence of triethylamine. All the synthesized conjugates reveal antiproliferative properties against A431 (squamous skin) cancer cell line with potency higher than that of 5-fluorouracil. Many of the synthesized agents also exhibit promising antiproliferative properties against HCT116 (colon) cancer cell line, of which 5o and 5c are the most effective with 12.9, 9.8 folds potency compared with Sunitinib. Promising activity is also shown against MCF7 (breast) cancer cell line with 1.19, 1.12 folds relative to 5-fluorouracil. PI-flow cytometry of compound 5c supports the arrest of cell cycle at G1-phase. However, compound 5o and Sunitinib arrest the cell cycle at S-phase. The synthesized conjugates can be considered as multi-targeted tyrosine kinase inhibitors due to the promising properties against VEGFR-2 and EGFR in MCF7 and HCT116. CDOCKER studies support the EGFR inhibitory properties. Compounds 5p and 5i possessing thienylidene heterocycle are anti-SARS-CoV-2 with high therapeutic indices. Many of the synthesized agents show enhanced COX-1/2 properties than aspirin with better selectivity index towards COX-2 relative to COX-1. The possible applicability of the potent candidates discovered as antitumor and anti-SARS-CoV-2 is supported by the safe profile against normal (non-cancer, RPE1 and VERO-E6) cells.
Collapse
Affiliation(s)
- Aladdin M. Srour
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Siva S. Panda
- Department of Chemistry & Physics, Augusta University, Augusta, GA 30912, US
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - May A. El-Manawaty
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Ahmed A. F. Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Ahmed El Taweel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | | | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt,Corresponding author
| |
Collapse
|
12
|
Ribone SR, Paz SA, Abrams CF, Villarreal MA. Target identification for repurposed drugs active against SARS-CoV-2 via high-throughput inverse docking. J Comput Aided Mol Des 2021; 36:25-37. [PMID: 34825285 PMCID: PMC8616721 DOI: 10.1007/s10822-021-00432-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022]
Abstract
Screening already approved drugs for activity against a novel pathogen can be an important part of global rapid-response strategies in pandemics. Such high-throughput repurposing screens have already identified several existing drugs with potential to combat SARS-CoV-2. However, moving these hits forward for possible development into drugs specifically against this pathogen requires unambiguous identification of their corresponding targets, something the high-throughput screens are not typically designed to reveal. We present here a new computational inverse-docking protocol that uses all-atom protein structures and a combination of docking methods to rank-order targets for each of several existing drugs for which a plurality of recent high-throughput screens detected anti-SARS-CoV-2 activity. We demonstrate validation of this method with known drug-target pairs, including both non-antiviral and antiviral compounds. We subjected 152 distinct drugs potentially suitable for repurposing to the inverse docking procedure. The most common preferential targets were the human enzymes TMPRSS2 and PIKfyve, followed by the viral enzymes Helicase and PLpro. All compounds that selected TMPRSS2 are known serine protease inhibitors, and those that selected PIKfyve are known tyrosine kinase inhibitors. Detailed structural analysis of the docking poses revealed important insights into why these selections arose, and could potentially lead to more rational design of new drugs against these targets.
Collapse
Affiliation(s)
- Sergio R Ribone
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), X5000HUA, Córdoba, Argentina
| | - S Alexis Paz
- Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , X5000HUA, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Fisicoquímica de Córdoba (INFIQC), X5000HUA, Córdoba, Argentina
| | - Cameron F Abrams
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Marcos A Villarreal
- Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba , X5000HUA, Córdoba, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Instituto de Fisicoquímica de Córdoba (INFIQC), X5000HUA, Córdoba, Argentina.
| |
Collapse
|
13
|
Price S, Bender SG, Yahn R, Till NA, Varady S, LaLonde RL. Searching for an ideal SERM: Mining tamoxifen structure-activity relationships. Bioorg Med Chem Lett 2021; 52:128383. [PMID: 34592434 DOI: 10.1016/j.bmcl.2021.128383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/08/2021] [Accepted: 09/21/2021] [Indexed: 11/15/2022]
Abstract
The repurposing of old drugs for new treatments has recently garnered increased attention in the face of new diseases and declining productivity of the pharmaceutial industry. This report draws attention to potential opportunities hiding in plain sight within the SAR of off-patent drugs. Herein we explore the untapped potential of Selective Estrogen Receptor Modulators (SERMs). SERMs are a class of molecules that have been highly influential in the treatment of estrogen receptor-positive breast cancers. However, the most commonly prescribed SERM, tamoxifen, has been found to increase the risk of endometrial cancer. Another SERM, raloxifene, does not increase incidence of endometrial cancer, but has been abandoned as a breast cancer treatment. We report the design, synthesis, and evaluation of an unexplored tamoxifen substitution pattern which mimics the geometry of raloxifene to confer its favorable pharmacodynamics. This substitution pattern was found to maintain excellent binding affinity to estrogen receptor-α.
Collapse
Affiliation(s)
- Sky Price
- Chemistry Department, Reed College, 3203 SE Woodstock Blvd, Portland, OR, USA; Department of Chemistry, University of Texas at Austin, 2506 Speedway, Austin, TX USA
| | - Sophie G Bender
- Chemistry Department, Reed College, 3203 SE Woodstock Blvd, Portland, OR, USA; Department of Chemistry and Chemical Biology, Cornell University, 122 Baker Laboratory, Ithaca, NY, USA
| | - Rachel Yahn
- Chemistry Department, Reed College, 3203 SE Woodstock Blvd, Portland, OR, USA
| | - Nicholas A Till
- Chemistry Department, Reed College, 3203 SE Woodstock Blvd, Portland, OR, USA
| | - Sophia Varady
- Chemistry Department, Reed College, 3203 SE Woodstock Blvd, Portland, OR, USA
| | - Rebecca Lyn LaLonde
- Chemistry Department, Reed College, 3203 SE Woodstock Blvd, Portland, OR, USA.
| |
Collapse
|
14
|
Abd-Alla HI, Souguir D, Radwan MO. Genus Sophora: a comprehensive review on secondary chemical metabolites and their biological aspects from past achievements to future perspectives. Arch Pharm Res 2021; 44:903-986. [PMID: 34907492 PMCID: PMC8671057 DOI: 10.1007/s12272-021-01354-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/29/2021] [Indexed: 12/13/2022]
Abstract
Sophora is deemed as one of the most remarkable genera of Fabaceae, and the third largest family of flowering plants. The genus Sophora comprises approximately 52 species, 19 varieties, and 7 forms that are widely distributed in Asia and mildly in Africa. Sophora species are recognized to be substantial sources of broad spectrum biopertinent secondary metabolites namely flavonoids, isoflavonoids, chalcones, chromones, pterocarpans, coumarins, benzofuran derivatives, sterols, saponins (mainly triterpene glycosides), oligostilbenes, and mainly alkaloids. Meanwhile, extracts and isolated compounds from Sophora have been identified to possess several health-promising effects including anti-inflammatory, anti-arthritic, antiplatelets, antipyretic, anticancer, antiviral, antimicrobial, antioxidant, anti-osteoporosis, anti-ulcerative colitis, antidiabetic, anti-obesity, antidiarrheal, and insecticidal activities. Herein, the present review aims to provide comprehensive details about the phytochemicals and biological effects of Sophora species. The review spotlighted on the promising phytonutrients extracted from Sophora and their plethora of bioactivities. The review also clarifies the remaining gaps and thus qualifies and supplies a platform for further investigations of these compounds.
Collapse
Affiliation(s)
- Howaida I Abd-Alla
- Chemistry of Natural Compounds Department, National Research Centre, El-Bohouth Street, Giza-Dokki, 12622, Egypt.
| | - Dalila Souguir
- Institut National de Recherches en Génie Rural, Eaux et Forêts (INRGREF), Université de Carthage, 10 Rue Hédi Karray, Manzeh IV, 2080, Ariana, Tunisia
| | - Mohamed O Radwan
- Chemistry of Natural Compounds Department, National Research Centre, El-Bohouth Street, Giza-Dokki, 12622, Egypt.
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
| |
Collapse
|
15
|
Disulfiram use is associated with lower risk of COVID-19: A retrospective cohort study. PLoS One 2021; 16:e0259061. [PMID: 34710137 PMCID: PMC8553043 DOI: 10.1371/journal.pone.0259061] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/11/2021] [Indexed: 12/02/2022] Open
Abstract
Effective, low-cost therapeutics are needed to prevent and treat COVID-19. Severe COVID-19 disease is linked to excessive inflammation. Disulfiram is an approved oral drug used to treat alcohol use disorder that is a potent anti-inflammatory agent and an inhibitor of the viral proteases. We investigated the potential effects of disulfiram on SARS-CoV-2 infection and disease severity in an observational study using a large database of clinical records from the national US Veterans Affairs healthcare system. A multivariable Cox regression adjusted for demographic information and diagnosis of alcohol use disorder revealed a reduced risk of SARS-CoV-2 infection with disulfiram use at a hazard ratio of 0.66 (34% lower risk, 95% confidence interval 24–43%). There were no COVID-19 related deaths among the 188 SARS-CoV-2 positive patients treated with disulfiram, in contrast to 5–6 statistically expected deaths based on the untreated population (P = 0.03). Our epidemiological results suggest that disulfiram may contribute to the reduced incidence and severity of COVID-19. These results support carefully planned clinical trials to assess the potential therapeutic effects of disulfiram in COVID-19.
Collapse
|
16
|
Revilla Pacheco C, Terán Hilares R, Colina Andrade G, Mogrovejo-Valdivia A, Pacheco Tanaka DA. Emerging contaminants, SARS-COV-2 and wastewater treatment plants, new challenges to confront: A short review. BIORESOURCE TECHNOLOGY REPORTS 2021; 15:100731. [PMID: 34124614 PMCID: PMC8183098 DOI: 10.1016/j.biteb.2021.100731] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022]
Abstract
The current pandemic caused by SARS-CoV-2 has put public health at risk, being wastewater-based epidemiology (WBE) a potential tool in the detection, prevention, and treatment of present and possible future outbreaks, since this virus enters wastewater through various sources such as feces, vomit, and sputum. Thus, advanced technologies such as advanced oxidation processes (AOP), membrane technology (MT) are identified through a systematic literature review as an alternative option for the destruction and removal of emerging contaminants (drugs and personal care products) released mainly by infected patients. The objectives of this review are to know the implications that the new COVID-19 outbreak is generating and will generate in water compartments, as well as the new challenges faced by wastewater treatment plants due to the change in a load of contaminants and the solutions proposed based on the aforementioned technologies to be applied to preserve public health and the environment.
Collapse
Affiliation(s)
- Claudia Revilla Pacheco
- Laboratorio de Tecnología de Membranas, Universidad Católica de Santa María - UCSM, Urb. San José, San José S/N, Yanahuara, Arequipa, Peru
| | - Ruly Terán Hilares
- Laboratorio de Tecnología de Membranas, Universidad Católica de Santa María - UCSM, Urb. San José, San José S/N, Yanahuara, Arequipa, Peru
| | - Gilberto Colina Andrade
- Laboratorio de Tecnología de Membranas, Universidad Católica de Santa María - UCSM, Urb. San José, San José S/N, Yanahuara, Arequipa, Peru
| | - Alejandra Mogrovejo-Valdivia
- Laboratorio de Tecnología de Membranas, Universidad Católica de Santa María - UCSM, Urb. San José, San José S/N, Yanahuara, Arequipa, Peru
| | - David Alfredo Pacheco Tanaka
- Laboratorio de Tecnología de Membranas, Universidad Católica de Santa María - UCSM, Urb. San José, San José S/N, Yanahuara, Arequipa, Peru
| |
Collapse
|
17
|
Spini A, Giudice V, Brancaleone V, Morgese MG, De Francia S, Filippelli A, Ruggieri A, Ziche M, Ortona E, Cignarella A, Trabace L. Sex-tailored pharmacology and COVID-19: Next steps towards appropriateness and health equity. Pharmacol Res 2021; 173:105848. [PMID: 34454035 PMCID: PMC8387562 DOI: 10.1016/j.phrs.2021.105848] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/21/2021] [Accepted: 08/22/2021] [Indexed: 12/12/2022]
Abstract
Making gender bias visible allows to fill the gaps in knowledge and understand health records and risks of women and men. The coronavirus disease 2019 (COVID-19) pandemic has shown a clear gender difference in health outcomes. The more severe symptoms and higher mortality in men as compared to women are likely due to sex and age differences in immune responses. Age-associated decline in sex steroid hormone levels may mediate proinflammatory reactions in older adults, thereby increasing their risk of adverse outcomes, whereas sex hormones and/or sex hormone receptor modulators may attenuate the inflammatory response and provide benefit to COVID-19 patients. While multiple pharmacological options including anticoagulants, glucocorticoids, antivirals, anti-inflammatory agents and traditional Chinese medicine preparations have been tested to treat COVID-19 patients with varied levels of evidence in terms of efficacy and safety, information on sex-targeted treatment strategies is currently limited. Women may have more benefit from COVID-19 vaccines than men, despite the occurrence of more frequent adverse effects, and long-term safety data with newly developed vectors are eagerly awaited. The prevalent inclusion of men in randomized clinical trials (RCTs) with subsequent extrapolation of results to women needs to be addressed, as reinforcing sex-neutral claims into COVID-19 research may insidiously lead to increased inequities in health care. The huge worldwide effort with over 3000 ongoing RCTs of pharmacological agents should focus on improving knowledge on sex, gender and age as pillars of individual variation in drug responses and enforce appropriateness.
Collapse
Affiliation(s)
- Andrea Spini
- University of Siena, Department of Medicine, Surgery and Neuroscience, 53100 Siena, Italy; University of Bordeaux, Bordeaux Population Health Center, UMR 1219, 33000 Bordeaux, France
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Vincenzo Brancaleone
- Department of Science, University of Basilicata, via Ateneo Lucano, 85100 Potenza, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Silvia De Francia
- Department of Clinical and Biological Sciences, S. Luigi Hospital, University of Turin, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Anna Ruggieri
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Ziche
- University of Siena, Department of Medicine, Surgery and Neuroscience, 53100 Siena, Italy; University of Bordeaux, Bordeaux Population Health Center, UMR 1219, 33000 Bordeaux, France; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Elena Ortona
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Andrea Cignarella
- Department of Medicine, University of Padova, via Giustiniani 2, 35128 Padova, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy; Centro Studi Nazionale Salute e Medicina di Genere, Italy.
| |
Collapse
|
18
|
de Melo GD, Lazarini F, Larrous F, Feige L, Kornobis E, Levallois S, Marchio A, Kergoat L, Hardy D, Cokelaer T, Pineau P, Lecuit M, Lledo P, Changeux J, Bourhy H. Attenuation of clinical and immunological outcomes during SARS-CoV-2 infection by ivermectin. EMBO Mol Med 2021; 13:e14122. [PMID: 34170074 PMCID: PMC8350903 DOI: 10.15252/emmm.202114122] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
The devastating pandemic due to SARS-CoV-2 and the emergence of antigenic variants that jeopardize the efficacy of current vaccines create an urgent need for a comprehensive understanding of the pathophysiology of COVID-19, including the contribution of inflammation to disease. It also warrants for the search of immunomodulatory drugs that could improve disease outcome. Here, we show that standard doses of ivermectin (IVM), an anti-parasitic drug with potential immunomodulatory activities through the cholinergic anti-inflammatory pathway, prevent clinical deterioration, reduce olfactory deficit, and limit the inflammation of the upper and lower respiratory tracts in SARS-CoV-2-infected hamsters. Whereas it has no effect on viral load in the airways of infected animals, transcriptomic analyses of infected lungs reveal that IVM dampens type I interferon responses and modulates several other inflammatory pathways. In particular, IVM dramatically reduces the Il-6/Il-10 ratio in lung tissue and promotes macrophage M2 polarization, which might account for the more favorable clinical presentation of IVM-treated animals. Altogether, this study supports the use of immunomodulatory drugs such as IVM, to improve the clinical condition of SARS-CoV-2-infected patients.
Collapse
Affiliation(s)
| | | | - Florence Larrous
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| | - Lena Feige
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| | - Etienne Kornobis
- Biomics Technological PlatformCenter for Technological Resources and Research (C2RT)Institut PasteurParisFrance
- Bioinformatics and Biostatistics HubComputational Biology DepartmentInstitut PasteurParisFrance
| | | | - Agnès Marchio
- Nuclear Organization and Oncogenesis UnitInstitut PasteurParisFrance
| | - Lauriane Kergoat
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| | - David Hardy
- Experimental Neuropathology UnitInstitut PasteurParisFrance
| | - Thomas Cokelaer
- Biomics Technological PlatformCenter for Technological Resources and Research (C2RT)Institut PasteurParisFrance
- Bioinformatics and Biostatistics HubComputational Biology DepartmentInstitut PasteurParisFrance
| | - Pascal Pineau
- Nuclear Organization and Oncogenesis UnitInstitut PasteurParisFrance
| | - Marc Lecuit
- Biology of Infection UnitInstitut PasteurInserm U1117ParisFrance
- Division of Infectious Diseases and Tropical MedicineInstitut ImagineUniversité de ParisNecker‐Enfants Malades University HospitalAP‐HPParisFrance
| | | | | | - Hervé Bourhy
- Lyssavirus Epidemiology and Neuropathology UnitInstitut PasteurParisFrance
| |
Collapse
|
19
|
Tsai KC, Lee YC, Tseng TS. Comprehensive Deep Mutational Scanning Reveals the Immune-Escaping Hotspots of SARS-CoV-2 Receptor-Binding Domain Targeting Neutralizing Antibodies. Front Microbiol 2021; 12:698365. [PMID: 34335530 PMCID: PMC8319916 DOI: 10.3389/fmicb.2021.698365] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
The rapid spread of SARS-CoV-2 has caused the COVID-19 pandemic, resulting in the collapse of medical care systems and economic depression worldwide. To combat COVID-19, neutralizing antibodies have been investigated and developed. However, the evolutions (mutations) of the receptor-binding domain (RBD) of SARS-CoV-2 enable escape from neutralization by these antibodies, further impairing recognition by the human immune system. Thus, it is critical to investigate and predict the putative mutations of RBD that escape neutralizing immune responses. Here, we employed computational analyses to comprehensively investigate the mutational effects of RBD on binding to neutralizing antibodies and angiotensin-converting enzyme 2 (ACE2) and demonstrated that the RBD residues K417, L452, L455, F456, E484, G485, F486, F490, Q493, and S494 were consistent with clinically emerging variants or experimental observations of attenuated neutralizations. We also revealed common hotspots, Y449, L455, and Y489, that exerted comparable destabilizing effects on binding to both ACE2 and neutralizing antibodies. Our results provide valuable information on the putative effects of RBD variants on interactions with neutralizing antibodies. These findings provide insights into possible evolutionary hotspots that can escape recognition by these antibodies. In addition, our study results will benefit the development and design of vaccines and antibodies to combat the newly emerging variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Lee
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- TMU Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Tien-Sheng Tseng
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
20
|
Klapars A, Chung JYL, Limanto J, Calabria R, Campeau LC, Campos KR, Chen W, Dalby SM, Davis TA, DiRocco DA, Hyde AM, Kassim AM, Larsen MU, Liu G, Maligres PE, Moment A, Peng F, Ruck RT, Shevlin M, Simmons BL, Song ZJ, Tan L, Wright TJ, Zultanski SL. Efficient synthesis of antiviral agent uprifosbuvir enabled by new synthetic methods. Chem Sci 2021; 12:9031-9036. [PMID: 34276931 PMCID: PMC8261776 DOI: 10.1039/d1sc01978c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/17/2021] [Indexed: 12/03/2022] Open
Abstract
An efficient route to the HCV antiviral agent uprifosbuvir was developed in 5 steps from readily available uridine in 50% overall yield. This concise synthesis was achieved by development of several synthetic methods: (1) complexation-driven selective acyl migration/oxidation; (2) BSA-mediated cyclization to anhydrouridine; (3) hydrochlorination using FeCl3/TMDSO; (4) dynamic stereoselective phosphoramidation using a chiral nucleophilic catalyst. The new route improves the yield of uprifosbuvir 50-fold over the previous manufacturing process and expands the tool set available for synthesis of antiviral nucleotides. An efficient route to the HCV antiviral agent uprifosbuvir was developed in 5 steps from readily available uridine in 50% overall yield.![]()
Collapse
Affiliation(s)
- Artis Klapars
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - John Y L Chung
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - John Limanto
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Ralph Calabria
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Louis-Charles Campeau
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Kevin R Campos
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Wenyong Chen
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Stephen M Dalby
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Tyler A Davis
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Daniel A DiRocco
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Alan M Hyde
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Amude M Kassim
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Mona Utne Larsen
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Guiquan Liu
- WuXi STA 90 Delin Road, Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Peter E Maligres
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Aaron Moment
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Feng Peng
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Rebecca T Ruck
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Michael Shevlin
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Bryon L Simmons
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Zhiguo Jake Song
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Lushi Tan
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Timothy J Wright
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| | - Susan L Zultanski
- Department of Process Research and Development, Merck & Co., Inc. Rahway New Jersey 07065 USA
| |
Collapse
|