51
|
Chang YJ, Yeh CY, Cheng JC, Huang YQ, Hsu KC, Lin YF, Lu CH. Potent sialic acid inhibitors that target influenza A virus hemagglutinin. Sci Rep 2021; 11:8637. [PMID: 33883588 PMCID: PMC8060387 DOI: 10.1038/s41598-021-87845-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/06/2021] [Indexed: 01/22/2023] Open
Abstract
Eradicating influenza A virus (IAV) is difficult, due to its genetic drift and reassortment ability. As the infectious cycle is initiated by the influenza glycoprotein, hemagglutinin (HA), which mediates the binding of virions to terminal sialic acids moieties, HA is a tempting target of anti-influenza inhibitors. However, the complexity of the HA structure has prevented delineation of the structural characterization of the HA protein-ligand complex. Our computational strategy efficiently analyzed > 200,000 records of compounds held in the United States National Cancer Institute (NCI) database and identified potential HA inhibitors, by modeling the sialic acid (SA) receptor binding site (RBS) for the HA structure. Our modeling revealed that compound NSC85561 showed significant antiviral activity against the IAV H1N1 strain with EC50 values ranging from 2.31 to 2.53 µM and negligible cytotoxicity (CC50 > 700 µM). Using the NSC85561 compound as the template to generate 12 derivatives, robust bioassay results revealed the strongest antiviral efficacies with NSC47715 and NSC7223. Virtual screening clearly identified three SA receptor binding site inhibitors that were successfully validated in experimental data. Thus, our computational strategy has identified SA receptor binding site inhibitors against HA that show IAV-associated antiviral activity.
Collapse
Affiliation(s)
- Yu-Jen Chang
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan
| | - Cheng-Yun Yeh
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Yu-Qi Huang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Yu-Feng Lin
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Chih-Hao Lu
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
52
|
Antivirals Targeting the Surface Glycoproteins of Influenza Virus: Mechanisms of Action and Resistance. Viruses 2021; 13:v13040624. [PMID: 33917376 PMCID: PMC8067422 DOI: 10.3390/v13040624] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/25/2022] Open
Abstract
Hemagglutinin and neuraminidase, which constitute the glycoprotein spikes expressed on the surface of influenza A and B viruses, are the most exposed parts of the virus and play critical roles in the viral lifecycle. As such, they make prominent targets for the immune response and antiviral drugs. Neuraminidase inhibitors, particularly oseltamivir, constitute the most commonly used antivirals against influenza viruses, and they have proved their clinical utility against seasonal and emerging influenza viruses. However, the emergence of resistant strains remains a constant threat and consideration. Antivirals targeting the hemagglutinin protein are relatively new and have yet to gain global use but are proving to be effective additions to the antiviral repertoire, with a relatively high threshold for the emergence of resistance. Here we review antiviral drugs, both approved for clinical use and under investigation, that target the influenza virus hemagglutinin and neuraminidase proteins, focusing on their mechanisms of action and the emergence of resistance to them.
Collapse
|
53
|
|
54
|
Sardar A, Lahiri A, Kamble M, Mallick AI, Tarafdar PK. Translation of Mycobacterium Survival Strategy to Develop a Lipo‐peptide based Fusion Inhibitor**. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202013848] [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)
- Avijit Sardar
- Department of Chemical Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Aritraa Lahiri
- Department of Biological Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Mithila Kamble
- Department of Biological Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Amirul I. Mallick
- Department of Biological Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| | - Pradip K. Tarafdar
- Department of Chemical Sciences Indian Institute of Science Education and Research Kolkata Mohanpur PIN-741246 India
| |
Collapse
|
55
|
Sardar A, Lahiri A, Kamble M, Mallick AI, Tarafdar PK. Translation of Mycobacterium Survival Strategy to Develop a Lipo-peptide based Fusion Inhibitor*. Angew Chem Int Ed Engl 2021; 60:6101-6106. [PMID: 33241871 PMCID: PMC7753697 DOI: 10.1002/anie.202013848] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Indexed: 12/16/2022]
Abstract
The entry of enveloped virus requires the fusion of viral and host cell membranes. An effective fusion inhibitor aiming at impeding such membrane fusion may emerge as a broad-spectrum antiviral agent against a wide range of viral infections. Mycobacterium survives inside the phagosome by inhibiting phagosome-lysosome fusion with the help of a coat protein coronin 1. Structural analysis of coronin 1 and other WD40-repeat protein suggest that the trp-asp (WD) sequence is placed at distorted β-meander motif (more exposed) in coronin 1. The unique structural feature of coronin 1 was explored to identify a simple lipo-peptide sequence (myr-WD), which effectively inhibits membrane fusion by modulating the interfacial order, water penetration, and surface potential. The mycobacterium inspired lipo-dipeptide was successfully tested to combat type 1 influenza virus (H1N1) and murine coronavirus infections as a potential broad-spectrum antiviral agent.
Collapse
Affiliation(s)
- Avijit Sardar
- Department of Chemical SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Aritraa Lahiri
- Department of Biological SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Mithila Kamble
- Department of Biological SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Amirul I. Mallick
- Department of Biological SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| | - Pradip K. Tarafdar
- Department of Chemical SciencesIndian Institute of Science Education and Research KolkataMohanpurPIN-741246India
| |
Collapse
|
56
|
Yang J, Zhang B, Huang Y, Liu T, Zeng B, Chai J, Wu J, Xu X. Antiviral activity and mechanism of ESC-1GN from skin secretion of hylarana guentheri against influenza a virus. J Biochem 2021; 169:757-765. [PMID: 33624755 DOI: 10.1093/jb/mvab019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/13/2021] [Indexed: 11/13/2022] Open
Abstract
Development of new and effective anti-influenza drugs is critical for prophylaxis and treatment of influenza A virus infection. A wide range of amphibian skin secretions have been identified to show antiviral activity. Our previously reported ESC-1GN, a peptide from the skin secretion of Hylarana guentheri, displayed good antimicrobial and anti-inflammatory effects. Here, we found that ESC-1GN possessed significant antiviral effects against influenza A viruses. Moreover, ESC-1GN could inhibit the entry of divergent H5N1 and H1N1 virus strains with the IC50 values from 1.29 to 4.59 μM. Mechanism studies demonstrated that ESC-1GN disrupted membrane fusion activity of influenza A viruses by interaction with HA2 subunit. The results of site-directed mutant assay and molecular docking revealed that E105, N50 and the residues around them on HA2 subunit could form hydrogen bonds with amino acid on ESC-1GN, which were critical for ESC-1GN binding to HA2 and inhibiting the entry of influenza A viruses. Altogether, these not only suggest that ESC-1GN maybe represent a new type of excellent template designing drugs against influenza A viruses, but also it may shed light on the immune mechanism and survival strategy of H. guentheri against viral pathogens.
Collapse
Affiliation(s)
- Jie Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bei Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yingna Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Teng Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Baishuang Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jingwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
57
|
Kocabiyik O, Cagno V, Silva PJ, Zhu Y, Sedano L, Bhide Y, Mettier J, Medaglia C, Da Costa B, Constant S, Huang S, Kaiser L, Hinrichs WLJ, Huckriede A, Le Goffic R, Tapparel C, Stellacci F. Non-Toxic Virucidal Macromolecules Show High Efficacy Against Influenza Virus Ex Vivo and In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001012. [PMID: 33552848 PMCID: PMC7856883 DOI: 10.1002/advs.202001012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 09/07/2020] [Indexed: 06/12/2023]
Abstract
Influenza is one of the most widespread viral infections worldwide and represents a major public health problem. The risk that one of the next pandemics is caused by an influenza strain is high. It is important to develop broad-spectrum influenza antivirals to be ready for any possible vaccine shortcomings. Anti-influenza drugs are available but they are far from ideal. Arguably, an ideal antiviral should target conserved viral domains and be virucidal, that is, irreversibly inhibit viral infectivity. Here, a new class of broad-spectrum anti-influenza macromolecules is described that meets these criteria and display exceedingly low toxicity. These compounds are based on a cyclodextrin core modified on its primary face with long hydrophobic linkers terminated either in 6'sialyl-N-acetyllactosamine (6'SLN) or in 3'SLN. SLN enables nanomolar inhibition of the viruses while the hydrophobic linkers confer irreversibility to the inhibition. The combination of these two properties allows for efficacy in vitro against several human or avian influenza strains, as well as against a 2009 pandemic influenza strain ex vivo. Importantly, it is shown that, in mice, one of the compounds provides therapeutic efficacy when administered 24 h post-infection allowing 90% survival as opposed to no survival for the placebo and oseltamivir.
Collapse
Affiliation(s)
- Ozgun Kocabiyik
- Insitute of MaterialsÉcole Polytechnique Fédérale de LausanneStation 12Lausanne1015Switzerland
| | - Valeria Cagno
- Insitute of MaterialsÉcole Polytechnique Fédérale de LausanneStation 12Lausanne1015Switzerland
- Department of Microbiology and Molecular MedicineUniversity of GenevaRue Michel Servet 1Geneva1205Switzerland
| | - Paulo Jacob Silva
- Insitute of MaterialsÉcole Polytechnique Fédérale de LausanneStation 12Lausanne1015Switzerland
| | - Yong Zhu
- Insitute of MaterialsÉcole Polytechnique Fédérale de LausanneStation 12Lausanne1015Switzerland
| | - Laura Sedano
- Virologie et Immunologie MoleculaireInstitut National Recherche AgronomiqueUniversité Paris‐SaclayJouy en Josas78350France
| | - Yoshita Bhide
- Department of Pharmaceutical Technology and BiopharmacyUniversity of GroningenGroningen9713GZThe Netherlands
- University Medical Center GroningenDepartment of Medical Microbiology and Infection Prevention (internal postcode EB88)University of GroningenHanzeplein 1Groningen9713GZThe Netherlands
| | - Joelle Mettier
- Virologie et Immunologie MoleculaireInstitut National Recherche AgronomiqueUniversité Paris‐SaclayJouy en Josas78350France
| | - Chiara Medaglia
- Department of Microbiology and Molecular MedicineUniversity of GenevaRue Michel Servet 1Geneva1205Switzerland
| | - Bruno Da Costa
- Virologie et Immunologie MoleculaireInstitut National Recherche AgronomiqueUniversité Paris‐SaclayJouy en Josas78350France
| | | | - Song Huang
- Epithelix SasChemin des Aulx 18Geneva1228Switzerland
| | - Laurent Kaiser
- Hopital Universitaire de GenèveRue Gabrielle Perret Gentil 4Geneva1205Switzerland
| | - Wouter L. J. Hinrichs
- Department of Pharmaceutical Technology and BiopharmacyUniversity of GroningenGroningen9713GZThe Netherlands
| | - Anke Huckriede
- University Medical Center GroningenDepartment of Medical Microbiology and Infection Prevention (internal postcode EB88)University of GroningenHanzeplein 1Groningen9713GZThe Netherlands
| | - Ronan Le Goffic
- Virologie et Immunologie MoleculaireInstitut National Recherche AgronomiqueUniversité Paris‐SaclayJouy en Josas78350France
| | - Caroline Tapparel
- Department of Microbiology and Molecular MedicineUniversity of GenevaRue Michel Servet 1Geneva1205Switzerland
| | - Francesco Stellacci
- Insitute of MaterialsÉcole Polytechnique Fédérale de LausanneStation 12Lausanne1015Switzerland
- Bioengineering InstituteEcole Polytechnique Fédérale de LausanneStation 12Lausanne1015Switzerland
| |
Collapse
|
58
|
Pillaiyar T, Manickam M, Meenakshisundaram S, Benjamine AJ. Candidate Drugs for the Potential Treatment of Coronavirus Diseases. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2021:85-114. [DOI: 10.1007/7653_2020_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
59
|
Nie C, Stadtmüller M, Parshad B, Wallert M, Ahmadi V, Kerkhoff Y, Bhatia S, Block S, Cheng C, Wolff T, Haag R. Heteromultivalent topology-matched nanostructures as potent and broad-spectrum influenza A virus inhibitors. SCIENCE ADVANCES 2021; 7:7/1/eabd3803. [PMID: 33523846 PMCID: PMC7775783 DOI: 10.1126/sciadv.abd3803] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/09/2020] [Indexed: 05/28/2023]
Abstract
Here, we report the topology-matched design of heteromultivalent nanostructures as potent and broad-spectrum virus entry inhibitors based on the host cell membrane. Initially, we investigate the virus binding dynamics to validate the better binding performance of the heteromultivalent moieties as compared to homomultivalent ones. The heteromultivalent binding moieties are transferred to nanostructures with a bowl-like shape matching the viral spherical surface. Unlike the conventional homomultivalent inhibitors, the heteromultivalent ones exhibit a half maximal inhibitory concentration of 32.4 ± 13.7 μg/ml due to the synergistic multivalent effects and the topology-matched shape. At a dose without causing cellular toxicity, >99.99% reduction of virus propagation has been achieved. Since multiple binding sites have also been identified on the S protein of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), we envision that the use of heteromultivalent nanostructures may also be applied to develop a potent inhibitor to prevent coronavirus infection.
Collapse
Affiliation(s)
- Chuanxiong Nie
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany
| | - Marlena Stadtmüller
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany
| | - Badri Parshad
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Matthias Wallert
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Vahid Ahmadi
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Yannic Kerkhoff
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Sumati Bhatia
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Stephan Block
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany.
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Thorsten Wolff
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany.
| | - Rainer Haag
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany.
| |
Collapse
|
60
|
Shao L, Yang F, Li W, Yu F. Design, Synthesis and Anti-influenza A Virus Evaluation of Oleanolic Acid C3-Glycoconjugates. CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202010029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
61
|
Terrier O, Slama-Schwok A. Anti-Influenza Drug Discovery and Development: Targeting the Virus and Its Host by All Possible Means. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1322:195-218. [PMID: 34258742 DOI: 10.1007/978-981-16-0267-2_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Infections by influenza virus constitute a major and recurrent threat for human health. Together with vaccines, antiviral drugs play a key role in the prevention and treatment of influenza virus infection and disease. Today, the number of antiviral molecules approved for the treatment of influenza is relatively limited, and their use is threatened by the emergence of viral strains with resistance mutations. There is therefore a real need to expand the prophylactic and therapeutic arsenal. This chapter summarizes the state of the art in drug discovery and development for the treatment of influenza virus infections, with a focus on both virus-targeting and host cell-targeting strategies. Novel antiviral strategies targeting other viral proteins or targeting the host cell, some of which are based on drug repurposing, may be used in combination to strengthen our therapeutic arsenal against this major pathogen.
Collapse
Affiliation(s)
- Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Anny Slama-Schwok
- Sorbonne Université, Centre de Recherche Saint-Antoine, INSERM U938, Biologie et Thérapeutique du Cancer, Paris, France.
| |
Collapse
|
62
|
Pillaiyar T, Wendt LL, Manickam M, Easwaran M. The recent outbreaks of human coronaviruses: A medicinal chemistry perspective. Med Res Rev 2021; 41:72-135. [PMID: 32852058 PMCID: PMC7461420 DOI: 10.1002/med.21724] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/22/2020] [Accepted: 08/08/2020] [Indexed: 01/18/2023]
Abstract
Coronaviruses (CoVs) infect both humans and animals. In humans, CoVs can cause respiratory, kidney, heart, brain, and intestinal infections that can range from mild to lethal. Since the start of the 21st century, three β-coronaviruses have crossed the species barrier to infect humans: severe-acute respiratory syndrome (SARS)-CoV-1, Middle East respiratory syndrome (MERS)-CoV, and SARS-CoV-2 (2019-nCoV). These viruses are dangerous and can easily be transmitted from human to human. Therefore, the development of anticoronaviral therapies is urgently needed. However, to date, no approved vaccines or drugs against CoV infections are available. In this review, we focus on the medicinal chemistry efforts toward the development of antiviral agents against SARS-CoV-1, MERS-CoV, SARS-CoV-2, targeting biochemical events important for viral replication and its life cycle. These targets include the spike glycoprotein and its host-receptors for viral entry, proteases that are essential for cleaving polyproteins to produce functional proteins, and RNA-dependent RNA polymerase for viral RNA replication.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal ChemistryUniversity of BonnBonnGermany
| | - Lukas L. Wendt
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal ChemistryUniversity of BonnBonnGermany
| | - Manoj Manickam
- Department of ChemistryPSG Institute of Technology and Applied ResearchCoimbatoreTamil NaduIndia
| | - Maheswaran Easwaran
- Department of Biomedical EngineeringSethu Institute of TechnologyVirudhunagarTamilnaduIndia
| |
Collapse
|
63
|
Do PC, Nguyen TH, Vo UHM, Le L. iBRAB: In silico based-designed broad-spectrum Fab against H1N1 influenza A virus. PLoS One 2020; 15:e0239112. [PMID: 33382708 PMCID: PMC7774956 DOI: 10.1371/journal.pone.0239112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/15/2020] [Indexed: 11/19/2022] Open
Abstract
Influenza virus A is a significant agent involved in the outbreak of worldwide epidemics, causing millions of fatalities around the world by respiratory diseases and seasonal illness. Many projects had been conducting to investigate recovered infected patients for therapeutic vaccines that have broad-spectrum activity. With the aid of the computational approach in biology, the designation for a vaccine model is more accessible. We developed an in silico protocol called iBRAB to design a broad-reactive Fab on a wide range of influenza A virus. The Fab model was constructed based on sequences and structures of available broad-spectrum Abs or Fabs against a wide range of H1N1 influenza A virus. As a result, the proposed Fab model followed iBRAB has good binding affinity over 27 selected HA of different strains of H1 influenza A virus, including wild-type and mutated ones. The examination also took by computational tools to fasten the procedure. This protocol could be applied for a fast-designed therapeutic vaccine against different types of threats.
Collapse
MESH Headings
- Amino Acid Sequence
- Antibodies, Viral/chemistry
- Antibodies, Viral/genetics
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Binding Sites
- Computer Simulation
- Drug Design
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/biosynthesis
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Molecular Docking Simulation
- Protein Binding
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- Protein Interaction Domains and Motifs
- Sequence Alignment
- Sequence Homology, Amino Acid
- Thermodynamics
Collapse
Affiliation(s)
- Phuc-Chau Do
- School of Biotechnology, International University, Thu Duc District, Hochiminh City, Vietnam
- Vietnam National University Ho Chi Minh City, Thu Duc District, Hochiminh City, Vietnam
| | - Trung H. Nguyen
- School of Biotechnology, International University, Thu Duc District, Hochiminh City, Vietnam
- Vietnam National University Ho Chi Minh City, Thu Duc District, Hochiminh City, Vietnam
| | - Uyen H. M. Vo
- School of Biotechnology, International University, Thu Duc District, Hochiminh City, Vietnam
- Vietnam National University Ho Chi Minh City, Thu Duc District, Hochiminh City, Vietnam
| | - Ly Le
- School of Biotechnology, International University, Thu Duc District, Hochiminh City, Vietnam
- Vietnam National University Ho Chi Minh City, Thu Duc District, Hochiminh City, Vietnam
- Vingroup Big Data Institute, Hai Ba Trung District, Ha Noi, Vietnam
| |
Collapse
|
64
|
Abd-Alla HI, Soltan MM, Hassan AZ, Taie HAA, Abo-Salem HM, Karam EA, El-Safty MM, Hanna AG. Cardenolides and pentacyclic triterpenes isolated from Acokanthera oblongifolia leaves: their biological activities with molecular docking study. ACTA ACUST UNITED AC 2020; 76:301-315. [PMID: 34218548 DOI: 10.1515/znc-2020-0198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/01/2020] [Indexed: 01/09/2023]
Abstract
Pentacyclic triterpenes and cardenolides were isolated from Acokanthera oblongifolia leaves. Their chemical structures were determined based on comprehensive 1D and 2D NMR spectroscopy. Their MIC was determined against 12 microorganisms. Their exerted cytotoxicity on the immortalized normal cells, hTERT-RPE1 was assessed by the sulforhodamine-B assay. The viral inhibitory effects of compounds against Newcastle disease virus (NDV) and H5N1 influenza virus IV were evaluated. Four in vitro antioxidant assays were performed in comparison with BHT and trolox and a weak activity was exhibited. Acovenoside A was with potent against H5N1-IV and NDV with IC50 ≤ 3.2 and ≤ 2.1 μg/ml and SI values of 93.75 and 95.23%, respectively, in comparison to ribavirin. Its CC50 record on Vero cells was > 400 and 200 μg/ml, respectively. Acobioside A was the most active compound against a broad range of microbes while Pseudomonas aeruginosa was the most sensitive. Its MIC (0.07 μg/ml) was 1/100-fold of the recorded CC50 (7.1 μg/ml/72 h) against hTERT-RPE1. The molecular docking of compounds on human DNA topoisomerase I (Top1-DNA) and IV glycoprotein hemagglutinin were studied using MOE program. This study has introduced the cardenolides rather than triterpenoids with the best docking score and binding interaction with the active site of the studied proteins.
Collapse
Affiliation(s)
- Howaida I Abd-Alla
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki-Giza, 12622, Egypt
| | - Maha M Soltan
- Chemistry of Medicinal Plants Department, Biology Unit, Central Laboratory for Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki-Giza, 12622, Egypt
| | - Amal Z Hassan
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki-Giza, 12622, Egypt
| | - Hanan A A Taie
- Plant Biochemistry Department, National Research Centre, Dokki-Giza, 12622, Egypt
| | - Heba M Abo-Salem
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki-Giza, 12622, Egypt
| | - Eman A Karam
- Microbial Chemistry Department, National Research Centre, Dokki-Giza, 12622, Egypt
| | - Mounir M El-Safty
- Central Laboratory for Evaluation of Veterinary Biologics, Abbassia-Cairo, 13181, Egypt
| | - Atef G Hanna
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki-Giza, 12622, Egypt
| |
Collapse
|
65
|
Zhang Q, Liang T, Nandakumar KS, Liu S. Emerging and state of the art hemagglutinin-targeted influenza virus inhibitors. Expert Opin Pharmacother 2020; 22:715-728. [PMID: 33327812 DOI: 10.1080/14656566.2020.1856814] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Introduction: Seasonal influenza vaccination, together with FDA-approved neuraminidase (NA) and polymerase acidic (PA) inhibitors, is the most effective way for prophylaxis and treatment of influenza infections. However, the low efficacy of prevailing vaccines to newly emerging influenza strains and increasing resistance to available drugs drives intense research to explore more effective inhibitors. Hemagglutinin (HA), one of the major surface proteins of influenza strains, represents an attractive therapeutic target to develop such new inhibitors.Areas covered: This review summarizes the current progress of HA-based influenza virus inhibitors and their mechanisms of action, which may facilitate further research in developing novel antiviral inhibitors for controlling influenza infections.Expert opinion: HA-mediated entry of influenza virus is an essential step for successful infection of the host, which makes HA a promising target for the development of antiviral drugs. Recent progress in delineating the crystal structures of HA, especially HA-inhibitors complexes, has revealed a number of key residues and conserved binding pockets within HA. This has opened up important insights for developing HA-based antiviral inhibitors that have a high resistance barrier and broad-spectrum activities.
Collapse
Affiliation(s)
- Qiao Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Taizhen Liang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Kutty Selva Nandakumar
- Southern Medical University-Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China.,State Key Laboratory of Organ Failure Research, Institute of Kidney Disease of Guangdong, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
66
|
Narayan C, Kwon J, Kim C, Kim SJ, Jang SK. Virus-based SELEX (viro-SELEX) allows development of aptamers targeting knotty proteins. Analyst 2020; 145:1473-1482. [PMID: 31868873 DOI: 10.1039/c9an01943j] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
It has been 100 years since the worst flu (Spanish flu) mankind has ever experienced. Rapid, accurate diagnosis and subtyping of flu are still an urgent unmet medical need. By using surrogate virus-based SELEX (viro-SELEX), we report here multiple advances incorporated into the field of flu diagnostics: (i) aptamers that can bind to the native virus well even though they cannot bind strongly to a recombinant protein (hemagglutinin); (ii) a couple of aptamers that can target a broad range of strains belonging to the H1N1 subtype and detect only the H1N1 subtype and nothing else; (iii) a highly sensitive lateral flow assay system (limit of detection is 0.08 HAU) using fluorescence-tagged aptamers. The viro-SELEX method of aptamer selection in conjunction with a fluorescent tag on aptamers is a very useful approach to develop highly sensitive, specific, portable, rapid, and quantitative point-of-care testing diagnostic tools for the future.
Collapse
Affiliation(s)
- Chandan Narayan
- Molecular virology Laboratory, Department of Life Sciences, Postech Biotech Center, Pohang University of Science and Technology, Cheongam-ro 77, Nam-gu, Pohang-si, Gyeongsangbuk-do 37673, Republic of Korea.
| | | | | | | | | |
Collapse
|
67
|
Li S, Jia X, Li H, Ye Y, Zhang X, Gao Y, Guo G, Liu S, Song G. Structure-aided optimization of 3-O-β-chacotriosyl epiursolic acid derivatives as novel H5N1 virus entry inhibitors. Bioorg Med Chem Lett 2020; 30:127518. [PMID: 32882419 DOI: 10.1016/j.bmcl.2020.127518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 01/16/2023]
Abstract
It is urgent to develop new antiviral agents due to the continuous emergence of drug-resistant strains of influenza virus. Our earlier studies have identified that certain pentacyclic triterpene saponins with 3-O-β-chacotriosyl residue are novel H5N1 virus entry inhibitors. In the present study, a series of C-28 modified 3-O-β-chacotriosyl epiursolic acid derivatives via conjugation with different kinds of sides were synthesized, of which anti-H5N1 activities in A549 cells were evaluated in vitro. Among them, 10 exhibited strongest anti-H5N1 potency at the low-micromole level without cytotoxicity, surpassing the potency of ribavirin. Further mechanism studies of the lead compound 10 based on HI, SPR and molecular modeling revealed that these new 3-epiursolic acid saponins could bind tightly to the viral envelope HA protein, thus blocking the invasion of H5N1 viruses into host cells.
Collapse
Affiliation(s)
- Sumei Li
- Department of Human Anatomy, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiuhua Jia
- Department of Ophthalmology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hui Li
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Yilu Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuesha Zhang
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Yongfeng Gao
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Guoqing Guo
- Department of Human Anatomy, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Shuwen Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Gaopeng Song
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
68
|
Xiu S, Dick A, Ju H, Mirzaie S, Abdi F, Cocklin S, Zhan P, Liu X. Inhibitors of SARS-CoV-2 Entry: Current and Future Opportunities. J Med Chem 2020; 63:12256-12274. [PMID: 32539378 PMCID: PMC7315836 DOI: 10.1021/acs.jmedchem.0c00502] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Indexed: 02/07/2023]
Abstract
Recently, a novel coronavirus initially designated 2019-nCoV but now termed SARS-CoV-2 has emerged and raised global concerns due to its virulence. SARS-CoV-2 is the etiological agent of "coronavirus disease 2019", abbreviated to COVID-19, which despite only being identified at the very end of 2019, has now been classified as a pandemic by the World Health Organization (WHO). At this time, no specific prophylactic or postexposure therapy for COVID-19 are currently available. Viral entry is the first step in the SARS-CoV-2 lifecycle and is mediated by the trimeric spike protein. Being the first stage in infection, entry of SARS-CoV-2 into host cells is an extremely attractive therapeutic intervention point. Within this review, we highlight therapeutic intervention strategies for anti-SARS-CoV, MERS-CoV, and other coronaviruses and speculate upon future directions for SARS-CoV-2 entry inhibitor designs.
Collapse
Affiliation(s)
- Siyu Xiu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| | - Han Ju
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Sako Mirzaie
- Department of Biochemistry, Sanandaj Branch, Islamic Azad University, Sanandaj 6616935391, Iran
| | - Fatemeh Abdi
- Department of Cellular and Molecular Biology, Islamic Azad University, Tehran North Branch, Tehran 1651153311, Iran
| | - Simon Cocklin
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| | - Peng Zhan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
69
|
A universal dual mechanism immunotherapy for the treatment of influenza virus infections. Nat Commun 2020; 11:5597. [PMID: 33154358 PMCID: PMC7645797 DOI: 10.1038/s41467-020-19386-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Seasonal influenza epidemics lead to 3–5 million severe infections and 290,000–650,000 annual global deaths. With deaths from the 1918 influenza pandemic estimated at >50,000,000 and future pandemics anticipated, the need for a potent influenza treatment is critical. In this study, we design and synthesize a bifunctional small molecule by conjugating the neuraminidase inhibitor, zanamivir, with the highly immunogenic hapten, dinitrophenyl (DNP), which specifically targets the surface of free virus and viral-infected cells. We show that this leads to simultaneous inhibition of virus release, and immune-mediated elimination of both free virus and virus-infected cells. Intranasal or intraperitoneal administration of a single dose of drug to mice infected with 100x MLD50 virus is shown to eradicate advanced infections from representative strains of both influenza A and B viruses. Since treatments of severe infections remain effective up to three days post lethal inoculation, our approach may successfully treat infections refractory to current therapies. In this study, the authors combine an anti-viral drug and immune system inducer to treat influenza A and B viral infections in vitro and in vivo. They show that the compound outperforms zanamivir alone as it is still able to clear infection three days post infection, and it can be administered via different routes without reduced efficacy.
Collapse
|
70
|
Ginex T, Luque FJ. Searching for effective antiviral small molecules against influenza A virus: A patent review. Expert Opin Ther Pat 2020; 31:53-66. [PMID: 33012213 DOI: 10.1080/13543776.2020.1831471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: Despite the current interest caused by SARS-Cov-2, influenza continues to be one of the most serious health concerns, with an estimated 1 billion cases across the globe, including 3-5 million severe cases and 290,000-650,000 deaths worldwide. Areas covered: This manuscript reviews the efforts made in the development of small molecules for the treatment of influenza virus, primarily focused on patent applications in the last 5 years. Attention is paid to compounds targeting key functional viral proteins, such as the M2 channel, neuraminidase, and hemagglutinin, highlighting the evolution toward new ligands and scaffolds motivated by the emergence of resistant strains. Finally, the discovery of compounds against novel viral targets, such as the RNA-dependent RNA polymerase, is discussed. Expert opinion: The therapeutic potential of antiviral agents is limited by the increasing presence of resistant strains. This should encourage research on novel strategies for therapeutic intervention. In this context, the discovery of arbidol and JNJ7918 against hemagglutinin, and current efforts on RNA-dependent RNA polymerase have disclosed novel opportunities for therapeutic treatment. Studies should attempt to expand the therapeutic arsenal of anti-flu agents, often in combined therapies, to prevent future health challenges caused by influenza virus. Abbreviations: AlphaLISA: amplified luminescent proximity homogeneous assay; HA: hemagglutinin; NA: neuraminidase; RBD: receptor binding domain; RdRp: RNA-dependent RNA polymerase; SA: sialic Acid; TBHQ: tert-butyl hydroquinone; TEVC: two-electrode voltage clamp.
Collapse
Affiliation(s)
- Tiziana Ginex
- Translational Medicinal and Biological Chemistry Group, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Biológicas (CIB-CSIC) , Madrid, Spain
| | - F Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona , Santa Coloma de Gramanet, Spain
| |
Collapse
|
71
|
Abdel-Aty AH, Khater MMA, Dutta H, Bouslimi J, Omri M. Computational solutions of the HIV-1 infection of CD4 + T-cells fractional mathematical model that causes acquired immunodeficiency syndrome (AIDS) with the effect of antiviral drug therapy. CHAOS, SOLITONS, AND FRACTALS 2020; 139:110092. [PMID: 32834626 PMCID: PMC7375326 DOI: 10.1016/j.chaos.2020.110092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/26/2020] [Accepted: 07/04/2020] [Indexed: 05/23/2023]
Abstract
This paper investigates the exact traveling wave solutions of the fractional model of the human immunodeficiency virus (HIV-1) infection for CD4 + T-cells. This model also treats with the effect of antiviral drug therapy. These solutions calculate both the boundary and initial conditions that allow employing the septic-B-spline scheme which is one of the most recent schemes in the numerical field. We use the obtained computational solutions via the modified Khater, the extended simplest equation, and sech-tanh methods through Atangana-Baleanu derivative operator. The comparison between the exact and numerical evaluated solutions is illustrated by some distinct sketches. The functioning of our numerical method is tested under three computational obtained solutions.
Collapse
Affiliation(s)
- Abdel-Haleem Abdel-Aty
- Department of Physics, College of Sciences, University of Bisha, P.O. Box 344, Bisha, 61922, Saudi Arabia
- Physics Department, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Mostafa M A Khater
- Department of Mathematics, Faculty of Science, Jiangsu University, Zhenjiang, 212013, China
- Department of Mathematics, Obour Institutes, Cairo, 11828, Egypt
| | - Hemen Dutta
- Department of Mathematics, Faculty of Science, Gauhati University, Guwahati 781014, India
| | - Jamel Bouslimi
- Department of Engineering Physics and Instrumentation, National Institute of Applied Sciences and Technology, Carthage University, Tunisia
- Physics Department, Faculty of Science, Taif University, PO.Box 888, 21974 Taif, Saudi Arabia
| | - M Omri
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
72
|
Structural Biology of Influenza Hemagglutinin: An Amaranthine Adventure. Viruses 2020; 12:v12091053. [PMID: 32971825 PMCID: PMC7551194 DOI: 10.3390/v12091053] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/27/2022] Open
Abstract
Hemagglutinin (HA) glycoprotein is an important focus of influenza research due to its role in antigenic drift and shift, as well as its receptor binding and membrane fusion functions, which are indispensable for viral entry. Over the past four decades, X-ray crystallography has greatly facilitated our understanding of HA receptor binding, membrane fusion, and antigenicity. The recent advances in cryo-EM have further deepened our comprehension of HA biology. Since influenza HA constantly evolves in natural circulating strains, there are always new questions to be answered. The incessant accumulation of knowledge on the structural biology of HA over several decades has also facilitated the design and development of novel therapeutics and vaccines. This review describes the current status of the field of HA structural biology, how we got here, and what the next steps might be.
Collapse
|
73
|
Yao Y, Kadam RU, Lee CCD, Woehl JL, Wu NC, Zhu X, Kitamura S, Wilson IA, Wolan DW. An influenza A hemagglutinin small-molecule fusion inhibitor identified by a new high-throughput fluorescence polarization screen. Proc Natl Acad Sci U S A 2020; 117:18431-18438. [PMID: 32690700 PMCID: PMC7414093 DOI: 10.1073/pnas.2006893117] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Influenza hemagglutinin (HA) glycoprotein is the primary surface antigen targeted by the host immune response and a focus for development of novel vaccines, broadly neutralizing antibodies (bnAbs), and therapeutics. HA enables viral entry into host cells via receptor binding and membrane fusion and is a validated target for drug discovery. However, to date, only a very few bona fide small molecules have been reported against the HA. To identity new antiviral lead candidates against the highly conserved fusion machinery in the HA stem, we synthesized a fluorescence-polarization probe based on a recently described neutralizing cyclic peptide P7 derived from the complementarity-determining region loops of human bnAbs FI6v3 and CR9114 against the HA stem. We then designed a robust binding assay compatible with high-throughput screening to identify molecules with low micromolar to nanomolar affinity to influenza A group 1 HAs. Our simple, low-cost, and efficient in vitro assay was used to screen H1/Puerto Rico/8/1934 (H1/PR8) HA trimer against ∼72,000 compounds. The crystal structure of H1/PR8 HA in complex with our best hit compound F0045(S) confirmed that it binds to pockets in the HA stem similar to bnAbs FI6v3 and CR9114, cyclic peptide P7, and small-molecule inhibitor JNJ4796. F0045 is enantioselective against a panel of group 1 HAs and F0045(S) exhibits in vitro neutralization activity against multiple H1N1 and H5N1 strains. Our assay, compound characterization, and small-molecule candidate should further stimulate the discovery and development of new compounds with unique chemical scaffolds and enhanced influenza antiviral capabilities.
Collapse
MESH Headings
- Antiviral Agents/chemistry
- Antiviral Agents/pharmacology
- Drug Evaluation, Preclinical/methods
- Fluorescence Polarization/methods
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Humans
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/metabolism
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/metabolism
- Influenza, Human/virology
- Small Molecule Libraries/chemistry
- Small Molecule Libraries/pharmacology
Collapse
Affiliation(s)
- Yao Yao
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Rameshwar U Kadam
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Chang-Chun David Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Jordan L Woehl
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Nicholas C Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Seiya Kitamura
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037;
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Dennis W Wolan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037;
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
74
|
Wu NC, Wilson IA. Influenza Hemagglutinin Structures and Antibody Recognition. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a038778. [PMID: 31871236 DOI: 10.1101/cshperspect.a038778] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hemagglutinin (HA) is most abundant glycoprotein on the influenza virus surface. Influenza HA promotes viral entry by engaging the receptor and mediating virus-host membrane fusion. At the same time, HA is the major antigen of the influenza virus. HA antigenic shift can result in pandemics, whereas antigenic drift allows human circulating strains to escape herd immunity. Most antibody responses against HA are strain-specific. However, antibodies that have neutralizing activities against multiple strains or even subtypes have now been discovered and characterized. These broadly neutralizing antibodies (bnAbs) target conserved regions on HA, such as the receptor-binding site and the stem domain. Structural studies of such bnAbs have provided important insight into universal influenza vaccine and therapeutic design. This review discusses the HA functions as well as HA-antibody interactions from a structural perspective.
Collapse
Affiliation(s)
- Nicholas C Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA.,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
75
|
Yu Y, Qin HJ, Shi XX, Song JQ, Zhou JP, Yu P, Fan ZC, Zhong M, Yang Y. Thiosialoside-decorated polymers use a two-step mechanism to inhibit both early and late stages of influenza virus infection. Eur J Med Chem 2020; 199:112357. [DOI: 10.1016/j.ejmech.2020.112357] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/16/2022]
|
76
|
Antanasijevic A, Durst MA, Cheng H, Gaisina IN, Perez JT, Manicassamy B, Rong L, Lavie A, Caffrey M. Structure of avian influenza hemagglutinin in complex with a small molecule entry inhibitor. Life Sci Alliance 2020; 3:3/8/e202000724. [PMID: 32611549 PMCID: PMC7335401 DOI: 10.26508/lsa.202000724] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
The binding properties of fusion inhibitor CBS1117 to hemagglutinin by x-ray crystallography, NMR, and mutagenesis give insights into mechanism and guidance for chemical optimization. HA plays a critical role in influenza infection and, thus HA is a potential target for antivirals. Recently, our laboratories have described a novel fusion inhibitor, termed CBS1117, with EC50 ∼3 μM against group 1 HA. In this work, we characterize the binding properties of CBS1117 to avian H5 HA by x-ray crystallography, NMR, and mutagenesis. The x-ray structure of the complex shows that the compound binds near the HA fusion peptide, a region that plays a critical role in HA-mediated fusion. NMR studies demonstrate binding of CBS1117 to H5 HA in solution and show extensive hydrophobic contacts between the compound and HA surface. Mutagenesis studies further support the location of the compound binding site proximal to the HA fusion peptide and identify additional amino acids that are important to compound binding. Together, this work gives new insights into the CBS1117 mechanism of action and can be exploited to further optimize this compound and better understand the group specific activity of small-molecule inhibitors of HA-mediated entry.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthew A Durst
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Han Cheng
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Jasmine T Perez
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | - Arnon Lavie
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
77
|
Wu G, Yu G, Yu Y, Yang S, Duan Z, Wang W, Liu Y, Yu R, Li J, Zhu T, Gu Q, Li D. Chemoreactive-Inspired Discovery of Influenza A Virus Dual Inhibitor to Block Hemagglutinin-Mediated Adsorption and Membrane Fusion. J Med Chem 2020; 63:6924-6940. [PMID: 32520560 DOI: 10.1021/acs.jmedchem.0c00312] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Owing to the emergence of drug resistance and high morbidity and mortality, the need for novel anti-influenza A virus (IAV) drugs with divergent targets is highly sought after. Herein, we reveal the discovery of an anti-IAV agent as a dual inhibitor to block hemagglutinin-mediated adsorption and membrane fusion using a chemoreactive ortho-quinone methide (o-QM) equivalent. Based on the o-QM equivalent nonenzymatically multipotent behavior, we created a series of clavatol-derived pseudo-natural products and found that penindolone (PND), a new diclavatol indole adduct, exhibited potent and broad-spectrum anti-IAV activities with low risk of inducing drug resistance. Distinct from current anti-IAV drugs, PND possesses a novel scaffold and is the first IAV inhibitor targeting both HA1 and HA2 subunits of virus hemagglutinin to dually block the IAV adsorption and membrane fusion process. More importantly, intranasal and oral administration of PND can protect mice against IAV-induced death and weight loss, superior to the effects of the clinical drug oseltamivir. Thus, the use of chemoreactive intermediates could expand our understanding of chemical diversity and aid in the development of anti-IAV drugs with novel targets.
Collapse
Affiliation(s)
- Guangwei Wu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Guihong Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Yunjia Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Shuang Yang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Zhongwei Duan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Wei Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, People's Republic of China
| | - Yankai Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Tianjiao Zhu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Qianqun Gu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China
| | - Dehai Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 26003, Shandong, P. R. China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, People's Republic of China
| |
Collapse
|
78
|
Tang Q, Xu Z, Jin M, Shu T, Chen Y, Feng L, Zhang Q, Lan K, Wu S, Zhou HB. Identification of dibucaine derivatives as novel potent enterovirus 2C helicase inhibitors: In vitro, in vivo, and combination therapy study. Eur J Med Chem 2020; 202:112310. [PMID: 32619885 DOI: 10.1016/j.ejmech.2020.112310] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/27/2020] [Accepted: 04/04/2020] [Indexed: 10/24/2022]
Abstract
Enterovirus A71 (EV-A71) is a human pathogen causing hand, foot and mouth disease (HFMD) which seriously threatened the safety and lives of infants and young children. However, there are no licensed direct antiviral agents to cure the HFMD. In this study, a series of quinoline formamide analogues as effective enterovirus inhibitors were developed, subsequent systematic structure-activity relationship (SAR) studies demonstrated that these quinoline formamide analogues exhibited good potency to treat EV-A71 infection. As described, the most efficient EV-A71 inhibitor 6i showed good anti-EV-A71 activity (EC50 = 1.238 μM) in RD cells. Furthermore, compound 6i could effectively prevent death of virus infected mice at dose of 6 mg/kg. When combined with emetine (0.1 mg/kg), this treatment could completely prevent the clinical symptoms and death of virus infected mice. Mechanism study indicated that compound 6i inhibited EV-A71 via targeting 2C helicase, thus impeding RNA remodeling and metabolism. Taken together, these data indicated that 6i is a promising EV-A71 inhibitor and worth extensive preclinical investigation as a lead compound.
Collapse
Affiliation(s)
- Qi Tang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Zhichao Xu
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Frontier Science Center for Immunology and Metabolism, Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Mengyu Jin
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Frontier Science Center for Immunology and Metabolism, Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, China
| | - Ting Shu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Yinuo Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Leilei Feng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Qiuhan Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China; Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| | - Shuwen Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Hai-Bing Zhou
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Frontier Science Center for Immunology and Metabolism, Wuhan University School of Pharmaceutical Sciences, Wuhan, 430071, China.
| |
Collapse
|
79
|
Weiss C, Carriere M, Fusco L, Capua I, Regla-Nava JA, Pasquali M, Scott JA, Vitale F, Unal MA, Mattevi C, Bedognetti D, Merkoçi A, Tasciotti E, Yilmazer A, Gogotsi Y, Stellacci F, Delogu LG. Toward Nanotechnology-Enabled Approaches against the COVID-19 Pandemic. ACS NANO 2020; 14:6383-6406. [PMID: 32519842 PMCID: PMC7299399 DOI: 10.1021/acsnano.0c03697] [Citation(s) in RCA: 354] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The COVID-19 outbreak has fueled a global demand for effective diagnosis and treatment as well as mitigation of the spread of infection, all through large-scale approaches such as specific alternative antiviral methods and classical disinfection protocols. Based on an abundance of engineered materials identifiable by their useful physicochemical properties through versatile chemical functionalization, nanotechnology offers a number of approaches to cope with this emergency. Here, through a multidisciplinary Perspective encompassing diverse fields such as virology, biology, medicine, engineering, chemistry, materials science, and computational science, we outline how nanotechnology-based strategies can support the fight against COVID-19, as well as infectious diseases in general, including future pandemics. Considering what we know so far about the life cycle of the virus, we envision key steps where nanotechnology could counter the disease. First, nanoparticles (NPs) can offer alternative methods to classical disinfection protocols used in healthcare settings, thanks to their intrinsic antipathogenic properties and/or their ability to inactivate viruses, bacteria, fungi, or yeasts either photothermally or via photocatalysis-induced reactive oxygen species (ROS) generation. Nanotechnology tools to inactivate SARS-CoV-2 in patients could also be explored. In this case, nanomaterials could be used to deliver drugs to the pulmonary system to inhibit interaction between angiotensin-converting enzyme 2 (ACE2) receptors and viral S protein. Moreover, the concept of "nanoimmunity by design" can help us to design materials for immune modulation, either stimulating or suppressing the immune response, which would find applications in the context of vaccine development for SARS-CoV-2 or in counteracting the cytokine storm, respectively. In addition to disease prevention and therapeutic potential, nanotechnology has important roles in diagnostics, with potential to support the development of simple, fast, and cost-effective nanotechnology-based assays to monitor the presence of SARS-CoV-2 and related biomarkers. In summary, nanotechnology is critical in counteracting COVID-19 and will be vital when preparing for future pandemics.
Collapse
Affiliation(s)
- Carsten Weiss
- Institute of Biological and Chemical
Systems, Biological Information Processing, Karlsruhe
Institute of Technology, Campus North,
Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen,
Germany
| | - Marie Carriere
- Univ. Grenoble
Alpes, CEA, CNRS, IRIG, SyMMES-CIBEST, F-38000
Grenoble, France
| | - Laura Fusco
- Department of Chemical and
Pharmaceutical Sciences, University of
Trieste, 34127 Trieste,
Italy
- Cancer Research Department,
Sidra Medicine, Doha,
Qatar
| | - Ilaria Capua
- One Health Center of Excellence,
University of Florida, Gainesville,
Florida 32611, United States
| | - Jose Angel Regla-Nava
- Division of Inflammation Biology,
La Jolla Institute for Allergy and
Immunology, La Jolla, California 92037,
United States
| | - Matteo Pasquali
- Department of Chemical &
Biomolecular Engineering, Rice University,
Houston, Texas 77251, United States
- Department of Chemistry,
Rice University, Houston, Texas
77251, United States
- Department of Materials Science and
Nanoengineering, Rice University, Houston,
Texas 77251, United States
| | - James A. Scott
- Dalla Lana School of Public Health,
University of Toronto, 223 College
Street, M5T 1R4 Toronto, Ontario, Canada
| | - Flavia Vitale
- Department of Neurology,
Bioengineering, Physical Medicine & Rehabilitation, Center for
Neuroengineering and Therapeutics, University of
Pennsylvania, Philadelphia, Pennsylvania 19104,
United States
- Center for Neurotrauma,
Neurodegeneration, and Restoration, Corporal Michael J.
Crescenz Veterans Affairs Medical Center,
Philadelphia, Pennsylvania 19104, United
States
| | | | - Cecilia Mattevi
- Department of Materials,
Imperial College London, London SW7
2AZ, United Kingdom
| | | | - Arben Merkoçi
- Nanobioelectronics & Biosensors
Group, Catalan Institute of Nanoscience and
Nanotechnology (ICN2), CSIC and BIST, Campus UAB,
08193 Bellaterra, Spain
- ICREA -
Institució Catalana de Recerca i Estudis
Avançats, ES-08010 Barcelona,
Spain
| | - Ennio Tasciotti
- Orthopedics and Sports Medicine,
Houston Methodist Hospital, Houston,
Texas 77030, United States
- Department of Plastic Surgery,
MD Anderson, Houston, Texas 77230,
United States
| | - Açelya Yilmazer
- Stem Cell Institute,
Ankara University, Ankara, 06100
Turkey
- Department of Biomedical Engineering,
Faculty of Engineering, Ankara University,
Ankara, 06100 Turkey
| | - Yury Gogotsi
- A.J. Drexel Nanomaterials Institute,
and Materials Science and Engineering Department, Drexel
University, Philadelphia, Pennsylvania 19104,
United States
| | - Francesco Stellacci
- Institute of Materials,
Ecole Polytechnique Federale de Lausanne
(EPFL), 1015 Lausanne,
Switzerland
- Interfaculty Bioengineering Institute,
Ecole Polytechnique Fédérale de
Lausanne (EPFL), 1015 Lausanne,
Switzerland
| | - Lucia Gemma Delogu
- Department of Biomedical Sciences,
University of Padua, 35122 Padova,
Italy
| |
Collapse
|
80
|
Nikolayeva YV, Ulashchik EA, Chekerda EV, Galochkina AV, Slesarchuk NA, Chistov AA, Nikitin TD, Korshun VA, Shmanai VV, Ustinov AV, Shtro AA. 5-(Perylen-3-ylethynyl)uracil Derivatives Inhibit Reproduction of Respiratory Viruses. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020; 46:315-320. [PMID: 32834709 PMCID: PMC7305479 DOI: 10.1134/s1068162020030139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 11/25/2019] [Accepted: 12/01/2019] [Indexed: 01/18/2023]
Abstract
In this work, we describe the synthesis of 5-(perylen-3-ylethynyl)uridine and its ability to effectively inhibit the replication of respiratory disease pathogens in cell culture, namely: influenza A virus (IVA); type 3 parainfluenza virus (PIV-3); and human respiratory syncytial virus (RSV). Related known compounds were also analyzed: 5-(perylen-3-ylethynyl)-2'-deoxy-uridine; 5-(perylen-3-ylethynyl)-arabino-uridine; and 1-carboxymethyl-3-pivaloyloxymethyl-5-(perylen-3-ylethynyl)uracil.
Collapse
Affiliation(s)
- Y. V. Nikolayeva
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| | - E. A. Ulashchik
- Institute of Physical Organic Chemistry of the NAS Belarus, 220072 Minsk, Belarus
| | - E. V. Chekerda
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| | - A. V. Galochkina
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| | - N. A. Slesarchuk
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Chemistry, Moscow State University, 119991 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
| | - A. A. Chistov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
| | - T. D. Nikitin
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Chemistry, Moscow State University, 119991 Moscow, Russia
| | - V. A. Korshun
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
- Gause Institute of New Antibiotics, 119021 Moscow, Russia
| | - V. V. Shmanai
- Institute of Physical Organic Chemistry of the NAS Belarus, 220072 Minsk, Belarus
| | - A. V. Ustinov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
- Department of Biology and Biotechnology, National Research University Higher School of Economics, 117312 Moscow, Russia
| | - A. A. Shtro
- Smorodintsev Research Institute of Influenza, 197376 St. Petersburg, Russia
| |
Collapse
|
81
|
Wu NC, Thompson AJ, Lee JM, Su W, Arlian BM, Xie J, Lerner RA, Yen HL, Bloom JD, Wilson IA. Different genetic barriers for resistance to HA stem antibodies in influenza H3 and H1 viruses. Science 2020; 368:1335-1340. [PMID: 32554590 PMCID: PMC7412937 DOI: 10.1126/science.aaz5143] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 04/14/2020] [Indexed: 12/19/2022]
Abstract
The discovery and characterization of broadly neutralizing human antibodies (bnAbs) to the highly conserved stem region of influenza hemagglutinin (HA) have contributed to considerations of a universal influenza vaccine. However, the potential for resistance to stem bnAbs also needs to be more thoroughly evaluated. Using deep mutational scanning, with a focus on epitope residues, we found that the genetic barrier to resistance to stem bnAbs is low for the H3 subtype but substantially higher for the H1 subtype owing to structural differences in the HA stem. Several strong resistance mutations in H3 can be observed in naturally circulating strains and do not reduce in vitro viral fitness and in vivo pathogenicity. This study highlights a potential challenge for development of a truly universal influenza vaccine.
Collapse
Affiliation(s)
- Nicholas C Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Andrew J Thompson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Juhye M Lee
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Wen Su
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Britni M Arlian
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jia Xie
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Richard A Lerner
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hui-Ling Yen
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jesse D Bloom
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
82
|
Yuan M, Liu H, Wu NC, Lee CCD, Zhu X, Zhao F, Huang D, Yu W, Hua Y, Tien H, Rogers TF, Landais E, Sok D, Jardine JG, Burton DR, Wilson IA. Structural basis of a public antibody response to SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.06.08.141267. [PMID: 32577642 PMCID: PMC7302194 DOI: 10.1101/2020.06.08.141267] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Molecular-level understanding of human neutralizing antibody responses to SARS-CoV-2 could accelerate vaccine design and facilitate drug discovery. We analyzed 294 SARS-CoV-2 antibodies and found that IGHV3-53 is the most frequently used IGHV gene for targeting the receptor binding domain (RBD) of the spike (S) protein. We determined crystal structures of two IGHV3-53 neutralizing antibodies +/- Fab CR3022 ranging from 2.33 to 3.11 Å resolution. The germline-encoded residues of IGHV3-53 dominate binding to the ACE2 binding site epitope with no overlap with the CR3022 epitope. Moreover, IGHV3-53 is used in combination with a very short CDR H3 and different light chains. Overall, IGHV3-53 represents a versatile public VH in neutralizing SARS-CoV-2 antibodies, where their specific germline features and minimal affinity maturation provide important insights for vaccine design and assessing outcomes.
Collapse
Affiliation(s)
- Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicholas C. Wu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chang-Chun D. Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Fangzhu Zhao
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Deli Huang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wenli Yu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yuanzi Hua
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Henry Tien
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas F. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Elise Landais
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI, New York, NY10004, USA
| | - Devin Sok
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI, New York, NY10004, USA
| | - Joseph G. Jardine
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI, New York, NY10004, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
83
|
Ethanol Extract of Caesalpinia decapetala Inhibits Influenza Virus Infection In Vitro and In Vivo. Viruses 2020; 12:v12050557. [PMID: 32443510 PMCID: PMC7290740 DOI: 10.3390/v12050557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022] Open
Abstract
Influenza virus infections can lead to viral pneumonia and acute respiratory distress syndrome in severe cases, causing significant morbidity and mortality and posing a great threat to human health. Because of the diversity of influenza virus strains and drug resistance to the current direct antiviral agents, there have been no effective drugs as yet to cure all patients infected by influenza viruses. Natural products from plants contain compounds with diverse structures that have the potential to interact with multiple host and virus factors. In this study, we identified the ethanol extract of Caesalpinia decapetala (Roth) Alston (EEC) as an inhibitor against the replication of a panel of influenza A and B viruses both on human pulmonary epithelial A549 and human monocytic U937 cells. The animal study revealed that EEC administration reduces the weight loss and improves the survival rate of mice infected with lethal influenza virus. Also, EEC treatment attenuated lung injury and reduced virus titer significantly. In conclusion, we showed that EEC has antiviral activity both in vitro and in vivo, suggesting that the plant C. decapetala has the potential to be further developed as a resource of new anti-influenza drugs.
Collapse
|
84
|
Hussein AFA, Cheng H, Tundup S, Antanasijevic A, Varhegyi E, Perez J, AbdulRahman EM, Elenany MG, Helal S, Caffrey M, Peet N, Manicassamy B, Rong L. Identification of entry inhibitors with 4-aminopiperidine scaffold targeting group 1 influenza A virus. Antiviral Res 2020; 177:104782. [PMID: 32222293 PMCID: PMC7243365 DOI: 10.1016/j.antiviral.2020.104782] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/20/2020] [Accepted: 03/22/2020] [Indexed: 01/09/2023]
Abstract
Influenza A viruses (IAVs) cause seasonal flu and occasionally pandemics. The current therapeutics against IAVs target two viral proteins - neuraminidase (NA) and M2 ion-channel protein. However, M2 ion channel inhibitors (amantadine and rimantadine) are no longer recommended by CDC for use due to the emergence of high level of antiviral resistance among the circulating influenza viruses, and resistant strains to NA inhibitors (oseltamivir and zanamivir) have also been reported. Therefore, development of novel anti-influenza therapies is urgently needed. As one of the viral surface glycoproteins, hemagglutinin (HA) mediates critical virus entry steps including virus binding to host cells and virus-host membrane fusion, which makes it a potential target for anti-influenza drug development. In this study, we report the identification of compound CBS1116 with a 4-aminopiperidine scaffold from a chemical library screen as an entry inhibitor specifically targeting two group 1 influenza A viruses, A/Puerto Rico/8/34 (H1N1) and recombinant low pathogenic avian H5N1 virus (A/Vietnam/1203/04, VN04Low). Mechanism of action studies show that CBS1116 interferes with the HA-mediated fusion process. Further structure activity relationship study generated a more potent compound CBS1117 which has a 50% inhibitory concentration of 70 nM and a selectivity index of ~4000 against A/Puerto Rico/8/34 (H1N1) infection in human lung epithelial cell line (A549).
Collapse
Affiliation(s)
- Amira F A Hussein
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Han Cheng
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Smanla Tundup
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, IL, 60439, USA; Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Elizabeth Varhegyi
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jasmine Perez
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Eiman M AbdulRahman
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mervat G Elenany
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Soheir Helal
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Norton Peet
- Chicago BioSolutions, Inc., 2242 West Harrison Suite 201, Chicago, IL, 60612, USA
| | - Balaji Manicassamy
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, IL, 60439, USA; Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
85
|
Aggarwal M, Plemper RK. Structural Insight into Paramyxovirus and Pneumovirus Entry Inhibition. Viruses 2020; 12:E342. [PMID: 32245118 PMCID: PMC7150754 DOI: 10.3390/v12030342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 01/04/2023] Open
Abstract
Paramyxoviruses and pneumoviruses infect cells through fusion (F) protein-mediated merger of the viral envelope with target membranes. Members of these families include a range of major human and animal pathogens, such as respiratory syncytial virus (RSV), measles virus (MeV), human parainfluenza viruses (HPIVs), and highly pathogenic Nipah virus (NiV). High-resolution F protein structures in both the metastable pre- and the postfusion conformation have been solved for several members of the families and a number of F-targeting entry inhibitors have progressed to advanced development or clinical testing. However, small-molecule RSV entry inhibitors have overall disappointed in clinical trials and viral resistance developed rapidly in experimental settings and patients, raising the question of whether the available structural information may provide a path to counteract viral escape through proactive inhibitor engineering. This article will summarize current mechanistic insight into F-mediated membrane fusion and examine the contribution of structural information to the development of small-molecule F inhibitors. Implications are outlined for future drug target selection and rational drug engineering strategies.
Collapse
Affiliation(s)
| | - Richard K Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA;
| |
Collapse
|
86
|
Gaisina IN, Peet NP, Cheng H, Li P, Du R, Cui Q, Furlong K, Manicassamy B, Caffrey M, Thatcher GRJ, Rong L. Optimization of 4-Aminopiperidines as Inhibitors of Influenza A Viral Entry That Are Synergistic with Oseltamivir. J Med Chem 2020; 63:3120-3130. [PMID: 32069052 DOI: 10.1021/acs.jmedchem.9b01900] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vaccination is the most prevalent prophylactic means for controlling seasonal influenza infections. However, an effective vaccine usually takes at least 6 months to develop for the circulating strains. Therefore, new therapeutic options are needed for the acute treatment of influenza infections to control this virus and prevent epidemics/pandemics from developing. We have discovered fast-acting, orally bioavailable acylated 4-aminopiperidines with an effective mechanism of action targeting viral hemagglutinin (HA). Our data show that these compounds are potent entry inhibitors of influenza A viruses. We present docking studies that suggest an HA binding site for these inhibitors on H5N1. Compound 16 displayed a significant decrease of viral titer when evaluated in the infectious assays with influenza virus H1N1 (A/Puerto Rico/8/1934) or H5N1 (A/Vietnam/1203/2004) strains and the oseltamivir-resistant strain with the most common H274Y mutation. In addition, compound 16 showed significant synergistic activity with oseltamivir in vitro.
Collapse
Affiliation(s)
- Irina N Gaisina
- UICentre (Drug Discovery@UIC) and Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States.,Chicago BioSolutions, Inc., 2242 West Harrison Street, Chicago, Illinois 60612, United States
| | - Norton P Peet
- Chicago BioSolutions, Inc., 2242 West Harrison Street, Chicago, Illinois 60612, United States
| | - Han Cheng
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 909 South Wolcott Avenue, Chicago, Illinois 60612, United States
| | - Ping Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 16369 Jinshi Road, Jinan, Shandong 250355, China
| | - Ruikun Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 16369 Jinshi Road, Jinan, Shandong 250355, China
| | - Qinghua Cui
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, 16369 Jinshi Road, Jinan, Shandong 250355, China
| | - Kevin Furlong
- Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, Illinois 60637, United States
| | - Balaji Manicassamy
- Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, Illinois 60637, United States.,Department of Microbiology and Immunology, University of Iowa, 51 Newton Road, Iowa City, Iowa 52242, United States
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| | - Gregory R J Thatcher
- UICentre (Drug Discovery@UIC) and Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 909 South Wolcott Avenue, Chicago, Illinois 60612, United States
| |
Collapse
|
87
|
An Oleanolic Acid Derivative Inhibits Hemagglutinin-Mediated Entry of Influenza A Virus. Viruses 2020; 12:v12020225. [PMID: 32085430 PMCID: PMC7077228 DOI: 10.3390/v12020225] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 01/09/2020] [Accepted: 02/14/2020] [Indexed: 01/09/2023] Open
Abstract
Influenza A viruses (IAV) have been a major public health threat worldwide, and options for antiviral therapy become increasingly limited with the emergence of drug-resisting virus strains. New and effective anti-IAV drugs, especially for highly pathogenic influenza, with different modes of action, are urgently needed. The influenza virus glycoprotein hemagglutinin (HA) plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a potential target for the development of anti-influenza drugs. In this study, we show that OA-10, a newly synthesized triterpene out of 11 oleanane-type derivatives, exhibited significant antiviral activity against four different subtypes of IAV (H1N1, H5N1, H9N2 and H3N2) replications in A549 cell cultures with EC50 ranging from 6.7 to 19.6 μM and a negligible cytotoxicity (CC50 > 640 μM). It inhibited acid-induced hemolysis in a dose-dependent manner, with an IC50 of 26 µM, and had a weak inhibition on the adsorption of H5 HA to chicken erythrocytes at higher concentrations (≥40 µM). Surface plasmon resonance (SPR) analysis showed that OA-10 interacted with HA in a dose-dependent manner with the equilibrium dissociation constants (KD) of the interaction of 2.98 × 10-12 M. Computer-aided molecular docking analysis suggested that OA-10 might bind to the cavity in HA stem region which is known to undergo significant rearrangement during membrane fusion. Our results demonstrate that OA-10 inhibits H5N1 IAV replication mainly by blocking the conformational changes of HA2 subunit required for virus fusion with endosomal membrane. These findings suggest that OA-10 could serve as a lead for further development of novel virus entry inhibitors to prevent and treat IAV infections.
Collapse
|
88
|
Díaz L, Soler D, Tresadern G, Buyck C, Perez-Benito L, Saen-Oon S, Guallar V, Soliva R. Monte Carlo simulations using PELE to identify a protein-protein inhibitor binding site and pose. RSC Adv 2020; 10:7058-7064. [PMID: 35493910 PMCID: PMC9049779 DOI: 10.1039/d0ra01127d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
In silico binding site location and pose prediction for a molecule targeted at a large protein surface is a challenging task. We report a blind test with two peptidomimetic molecules that bind the flu virus hemagglutinin (HA) surface antigen, JNJ7918 and JNJ4796 (recently disclosed in van Dongen et al., Science, 2019, 363). Tests with a series of conventional approaches such as rigid (receptor) docking against available X-ray crystal structures or against an ensemble of structures generated by quick methodologies (NMA, homology modeling) gave mixed results, due to the shallowness and flexibility of the binding site and the sheer size of the target. However, tests with our Monte Carlo platform PELE in two protocols involving either exploration of the whole protein surface (global exploration), or the latter followed by refinement of best solutions (local exploration) yielded remarkably good results by locating the actual binding site and generating binding modes that recovered all native contacts found in the X-ray structures. Thus, the Monte Carlo scheme of PELE seems promising as a quick methodology to overcome the challenge of identifying entirely unknown binding sites and modes for protein–protein disruptors. PELE prospectively unveils the binding site and mode of a protein–protein disruptor.![]()
Collapse
Affiliation(s)
- Lucía Díaz
- Nostrum Biodiscovery Jordi Girona 29, Nexus II D128 08034 Barcelona Spain
| | - Daniel Soler
- Nostrum Biodiscovery Jordi Girona 29, Nexus II D128 08034 Barcelona Spain
| | - Gary Tresadern
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. Turnhoutseweg 30, B-2340 Beerse Belgium
| | - Christophe Buyck
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. Turnhoutseweg 30, B-2340 Beerse Belgium
| | - Laura Perez-Benito
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica N. V. Turnhoutseweg 30, B-2340 Beerse Belgium
| | - Suwipa Saen-Oon
- Nostrum Biodiscovery Jordi Girona 29, Nexus II D128 08034 Barcelona Spain
| | - Victor Guallar
- Barcelona Supercomputing Center, Join IRB-BSC Program in Computational Biology Spain.,ICREA Passeig Lluís Companys 23 E-08010 Barcelona Spain
| | - Robert Soliva
- Nostrum Biodiscovery Jordi Girona 29, Nexus II D128 08034 Barcelona Spain
| |
Collapse
|
89
|
Hellert J, Buchrieser J, Larrous F, Minola A, de Melo GD, Soriaga L, England P, Haouz A, Telenti A, Schwartz O, Corti D, Bourhy H, Rey FA. Structure of the prefusion-locking broadly neutralizing antibody RVC20 bound to the rabies virus glycoprotein. Nat Commun 2020; 11:596. [PMID: 32001700 PMCID: PMC6992781 DOI: 10.1038/s41467-020-14398-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/02/2020] [Indexed: 10/25/2022] Open
Abstract
Rabies virus (RABV) causes fatal encephalitis in more than 59,000 people yearly. Upon the bite of an infected animal, the development of clinical disease can be prevented with post-exposure prophylaxis (PEP), which includes the administration of Rabies immunoglobulin (RIG). However, the high cost and limited availability of serum-derived RIG severely hamper its wide use in resource-limited countries. A safe low-cost alternative is provided by using broadly neutralizing monoclonal antibodies (bnAbs). Here we report the X-ray structure of one of the most potent and most broadly reactive human bnAbs, RVC20, in complex with its target domain III of the RABV glycoprotein (G). The structure reveals that the RVC20 binding determinants reside in a highly conserved surface of G, rationalizing its broad reactivity. We further show that RVC20 blocks the acid-induced conformational change required for membrane fusion. Our results may guide the future development of direct antiviral small molecules for Rabies treatment.
Collapse
Affiliation(s)
- Jan Hellert
- Structural Virology Unit, Institut Pasteur, CNRS UMR 3569, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France
| | - Julian Buchrieser
- Virus and Immunity Unit, Institut Pasteur, CNRS UMR 3569, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France
| | - Florence Larrous
- Lyssavirus Epidemiology and Neuropathology Unit, Institut Pasteur, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France
| | - Andrea Minola
- Humabs BioMed SA, a subsidiary of Vir Biotechnology Inc., Via dei Gaggini 3, 6500, Bellinzona, Switzerland
| | - Guilherme Dias de Melo
- Lyssavirus Epidemiology and Neuropathology Unit, Institut Pasteur, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France
| | - Leah Soriaga
- Vir Biotechnology Inc, San Francisco, CA, 94158, USA
| | - Patrick England
- Molecular Biophysics Platform C2RT, Institut Pasteur, CNRS UMR 3528, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France
| | - Ahmed Haouz
- Crystallography Platform C2RT, Institut Pasteur, CNRS UMR 3528, 25-28 rue du Dr. Roux, Cedex 15, 75724, Paris, France
| | | | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, CNRS UMR 3569, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France
| | - Davide Corti
- Humabs BioMed SA, a subsidiary of Vir Biotechnology Inc., Via dei Gaggini 3, 6500, Bellinzona, Switzerland
| | - Hervé Bourhy
- Lyssavirus Epidemiology and Neuropathology Unit, Institut Pasteur, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France.
| | - Félix A Rey
- Structural Virology Unit, Institut Pasteur, CNRS UMR 3569, 25-28 rue du Docteur Roux, Cedex 15, 75724, Paris, France.
| |
Collapse
|
90
|
Emetine protects mice from enterovirus infection by inhibiting viral translation. Antiviral Res 2020; 173:104650. [DOI: 10.1016/j.antiviral.2019.104650] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 11/19/2022]
|
91
|
Cihan-Üstündağ G, Zopun M, Vanderlinden E, Ozkirimli E, Persoons L, Çapan G, Naesens L. Superior inhibition of influenza virus hemagglutinin-mediated fusion by indole-substituted spirothiazolidinones. Bioorg Med Chem 2020; 28:115130. [DOI: 10.1016/j.bmc.2019.115130] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/12/2019] [Accepted: 09/18/2019] [Indexed: 12/26/2022]
|
92
|
Zhu X, Turner HL, Lang S, McBride R, Bangaru S, Gilchuk IM, Yu W, Paulson JC, Crowe JE, Ward AB, Wilson IA. Structural Basis of Protection against H7N9 Influenza Virus by Human Anti-N9 Neuraminidase Antibodies. Cell Host Microbe 2019; 26:729-738.e4. [PMID: 31757767 DOI: 10.1016/j.chom.2019.10.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/20/2019] [Accepted: 10/01/2019] [Indexed: 01/22/2023]
Abstract
Influenza virus neuraminidase (NA) is a major target for small-molecule antiviral drugs. Antibodies targeting the NA surface antigen could also inhibit virus entry and egress to provide host protection. However, our understanding of the nature and range of target epitopes is limited because of a lack of human antibody structures with influenza neuraminidase. Here, we describe crystal and cryogenic electron microscopy (cryo-EM) structures of NAs from human-infecting avian H7N9 viruses in complex with five human anti-N9 antibodies, systematically defining several antigenic sites and antibody epitope footprints. These antibodies either fully or partially block the NA active site or bind to epitopes distant from the active site while still showing neuraminidase inhibition. The inhibition of antibodies to NAs was further analyzed by glycan array and solution-based NA activity assays. Together, these structural studies provide insights into protection by anti-NA antibodies and templates for the development of NA-based influenza virus vaccines and therapeutics.
Collapse
Affiliation(s)
- Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hannah L Turner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shanshan Lang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ryan McBride
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Sandhya Bangaru
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Iuliia M Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Wenli Yu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
93
|
Li H, Chen L, Li S, Liao Y, Wang L, Liu Z, Liu S, Song G. Incorporation of privileged structures into 3-O-β-chacotriosyl ursolic acid can enhance inhibiting the entry of the H5N1 virus. Bioorg Med Chem Lett 2019; 29:2675-2680. [PMID: 31371135 DOI: 10.1016/j.bmcl.2019.07.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/09/2019] [Accepted: 07/18/2019] [Indexed: 01/27/2023]
Abstract
The glycoprotein hemagglutinin of influenza virus plays a key role in the initial stage of virus infection, making it a potential target for novel influenza viruses entry inhibitors. Two "privileged fragments", 2-(piperidin-1-yl)ethan-1-amine and 2-(1,3-oxazinan-3-yl)ethan-1-amine were integrated into 3-O-β-chacotriosyl ursolic acid producing new derivatives 5 and 6 with improved activity against IAVs in vitro. Mechanistically, compound 6 was effective in inhibiting infection of H1-, H3-, and H5-typed influenza A viruses by interfering with the viral hemagglutinin. Furthermore, the docking studies were in agreement with the antiviral data. These results showed that the title compound 6 as a new lead compound was meriting further optimization and development.
Collapse
Affiliation(s)
- Hui Li
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Lizhu Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sumei Li
- Department of Human Anatomy, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yixian Liao
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Integrative Microbiology Research Centre, Guangdong Province Key Laboratory of Microbial Signals and Disease Control, South China Agricultural University, Guangzhou 510642, China
| | - Lei Wang
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Zhihao Liu
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Shuwen Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Gaopeng Song
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
94
|
Abstract
The process of entry into a host cell is a key step in the life cycle of most viruses. In recent years, there has been a significant increase in our understanding of the routes and mechanisms of entry for a number of these viruses. This has led to the development of novel broad-spectrum antiviral approaches that target host cell proteins and pathways, in addition to strategies focused on individual viruses or virus families. Here we consider a number of these approaches and their broad-spectrum potential.
Collapse
Affiliation(s)
- Michela Mazzon
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Mark Marsh
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
95
|
Novel Small Molecule Targeting the Hemagglutinin Stalk of Influenza Viruses. J Virol 2019; 93:JVI.00878-19. [PMID: 31167918 DOI: 10.1128/jvi.00878-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/02/2019] [Indexed: 11/20/2022] Open
Abstract
Combating influenza is one of the perennial global public health issues to be managed. Antiviral drugs are useful for the treatment of influenza in the absence of an appropriate vaccine. However, the appearance of resistant strains necessitates a constant search for new drugs. In this study, we investigated novel anti-influenza drug candidates using in vitro and in vivo assays. We identified anti-influenza hit compounds using a high-throughput screening method with a green fluorescent protein-tagged recombinant influenza virus. Through subsequent analyses of their cytotoxicity and pharmacokinetic properties, one candidate (IY7640) was selected for further evaluation. In a replication kinetics analysis, IY7640 showed greater inhibitory effects during the early phase of viral infection than the viral neuraminidase inhibitor oseltamivir. In addition, we observed that hemagglutinin (HA)-mediated membrane fusion was inhibited by IY7640 treatment, indicating that the HA stalk region, which is highly conserved across various (sub)types of influenza viruses, may be the molecular target of IY7640. In an escape mutant analysis in cells, amino acid mutations were identified at the HA stalk region of the 2009 pandemic H1N1 (pH1N1) virus. Even though the in vivo efficacy of IY7640 did not reach complete protection in a lethal challenge study in mice, these results suggest that IY7640 has potential to be developed as a new type of anti-influenza drug.IMPORTANCE Anti-influenza drugs with broad-spectrum efficacy against antigenically diverse influenza viruses can be highly useful when no vaccines are available. To develop new anti-influenza drugs, we screened a number of small molecules and identified a strong candidate, IY7640. When added at the time of or after influenza virus infection, IY7640 was observed to successfully inhibit or reduce viral replication in cells. We subsequently discovered that IY7640 targets the stalk region of the influenza HA protein, which exhibits a relatively high degree of amino acid sequence conservation across various (sub)types of influenza viruses. Furthermore, IY7640 was observed to block HA-mediated membrane fusion of H1N1, H3N2, and influenza B viruses in cells. Although it appears less effective against strains other than H1N1 subtype viruses in a challenge study in mice, we suggest that the small molecule IY7640 has potential to be optimized as a new anti-influenza drug.
Collapse
|
96
|
A conserved histidine in Group-1 influenza subtype hemagglutinin proteins is essential for membrane fusion activity. Virology 2019; 536:78-90. [PMID: 31401467 DOI: 10.1016/j.virol.2019.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Abstract
Influenza A viruses enter host cells through the endocytic pathway, where acidification triggers conformational changes of the viral hemagglutinin (HA) to drive membrane fusion. During this process, the HA fusion peptide is extruded from its buried position in the neutral pH structure and targeted to the endosomal membrane. Conserved ionizable residues near the fusion peptide may play a role in initiating these structural rearrangements. We targeted highly conserved histidine residues in this region, at HA1 position 17 of Group-2 HA subtypes and HA2 position 111 of Group-1 HA subtypes, to determine their role in fusion activity. WT and mutant HA proteins representing several subtypes were expressed and characterized, revealing that His 111 is essential for HA functional activity of Group-1 subtypes, supporting continued efforts to target this region of the HA structure for vaccination strategies and the design of antiviral compounds.
Collapse
|
97
|
Zhang Y, Xu C, Zhang H, Liu GD, Xue C, Cao Y. Targeting Hemagglutinin: Approaches for Broad Protection against the Influenza A Virus. Viruses 2019; 11:v11050405. [PMID: 31052339 PMCID: PMC6563292 DOI: 10.3390/v11050405] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/26/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022] Open
Abstract
Influenza A viruses are dynamically epidemic and genetically diverse. Due to the antigenic drift and shift of the virus, seasonal vaccines are required to be reformulated annually to match with current circulating strains. However, the mismatch between vaccinal strains and circulating strains occurs frequently, resulting in the low efficacy of seasonal vaccines. Therefore, several “universal” vaccine candidates based on the structure and function of the hemagglutinin (HA) protein have been developed to meet the requirement of a broad protection against homo-/heterosubtypic challenges. Here, we review recent novel constructs and discuss several important findings regarding the broad protective efficacy of HA-based universal vaccines.
Collapse
Affiliation(s)
- Yun Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Cong Xu
- Research Center of Agricultural of Dongguan City, Dongguan 523086, China.
| | - Hao Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - George Dacai Liu
- Firstline Biopharmaceuticals Corporation, 12,050 167th PL NE, Redmond, WA 98052, USA.
| | - Chunyi Xue
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Yongchang Cao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
98
|
Cully M. Antibody-inspired small molecule for influenza protection. Nat Rev Drug Discov 2019:10.1038/d41573-019-00053-4. [PMID: 31048797 DOI: 10.1038/d41573-019-00053-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|