1
|
Chakraborty S, Chauhan A. Fighting the flu: a brief review on anti-influenza agents. Biotechnol Genet Eng Rev 2024; 40:858-909. [PMID: 36946567 DOI: 10.1080/02648725.2023.2191081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
The influenza virus causes one of the most prevalent and lethal infectious viral diseases of the respiratory system; the disease progression varies from acute self-limiting mild fever to disease chronicity and death. Although both the preventive and treatment measures have been vital in protecting humans against seasonal epidemics or sporadic pandemics, there are several challenges to curb the influenza virus such as limited or poor cross-protection against circulating virus strains, moderate protection in immune-compromised patients, and rapid emergence of resistance. Currently, there are four US-FDA-approved anti-influenza drugs to treat flu infection, viz. Rapivab, Relenza, Tamiflu, and Xofluza. These drugs are classified based on their mode of action against the viral replication cycle with the first three being Neuraminidase inhibitors, and the fourth one targeting the viral polymerase. The emergence of the drug-resistant strains of influenza, however, underscores the need for continuous innovation towards development and discovery of new anti-influenza agents with enhanced antiviral effects, greater safety, and improved tolerability. Here in this review, we highlighted commercially available antiviral agents besides those that are at different stages of development including under clinical trials, with a brief account of their antiviral mechanisms.
Collapse
Affiliation(s)
| | - Ashwini Chauhan
- Department of Microbiology, Tripura University, Agartala, India
| |
Collapse
|
2
|
Paparoditis PCG, Fruehwirth A, Bevc K, Low JS, Jerak J, Terzaghi L, Foglierini M, Fernandez B, Jarrossay D, Corti D, Sallusto F, Lanzavecchia A, Cassotta A. Site-specific serology unveils cross-reactive monoclonal antibodies targeting influenza A hemagglutinin epitopes. Eur J Immunol 2024; 54:e2451045. [PMID: 39031535 DOI: 10.1002/eji.202451045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024]
Abstract
Efficient identification of human monoclonal antibodies targeting specific antigenic sites is pivotal for advancing vaccines and immunotherapies against infectious diseases and cancer. Existing screening techniques, however, limit our ability to discover monoclonal antibodies with desired specificity. In this study, we introduce a novel method, blocking of binding (BoB) enzyme-linked immunoassay (ELISA), enabling the detection of high-avidity human antibodies directed to defined epitopes. Leveraging BoB-ELISA, we analyzed the antibody response to known epitopes of influenza A hemagglutinin (HA) in the serum of vaccinated donors. Our findings revealed that serum antibodies targeting head epitopes were immunodominant, whereas antibodies against the stem epitope, although subdominant, were highly prevalent. Extending our analysis across multiple HA strains, we examined the cross-reactive antibody response targeting the stem epitope. Importantly, employing BoB-ELISA we identified donors harboring potent heterosubtypic antibodies targeting the HA stem. B-cell clonal analysis of these donors revealed three novel, genealogically independent monoclonal antibodies with broad cross-reactivity to multiple HAs. In summary, we demonstrated that BoB-ELISA is a sensitive technique for measuring B-cell epitope immunogenicity, enabling the identification of novel monoclonal antibodies with implications for enhanced vaccine development and immunotherapies.
Collapse
Affiliation(s)
- Philipp C G Paparoditis
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Alexander Fruehwirth
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Kajetana Bevc
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Josipa Jerak
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Laura Terzaghi
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Mathilde Foglierini
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Blanca Fernandez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - David Jarrossay
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute for Microbiology, ETH Zurich, Zurich, Switzerland
| | - Antonio Lanzavecchia
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
- National Institute of Molecular Genetics, Milano, Italy
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
3
|
Franchini M, Focosi D. Monoclonal Antibodies and Hyperimmune Immunoglobulins in the Next Pandemic. Curr Top Microbiol Immunol 2024. [PMID: 38877202 DOI: 10.1007/82_2024_274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Pandemics are highly unpredictable events that are generally caused by novel viruses. There is a high likelihood that such novel pathogens belong to entirely novel viral families for which no targeted small-molecule antivirals exist. In addition, small-molecule antivirals often have pharmacokinetic properties that make them contraindicated for the frail patients who are often the most susceptible to a novel virus. Passive immunotherapies-available from the first convalescent patients-can then play a key role in controlling pandemics. Convalescent plasma is immediately available, but if manufacturers have fast platforms to generate marketable drugs, other forms of passive antibody treatment can be produced. In this chapter, we will review the technological platforms for generating monoclonal antibodies and hyperimmune immunoglobulins, the current experience on their use for treatment of COVID-19, and the pipeline for pandemic candidates.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantua, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy.
| |
Collapse
|
4
|
Li Y, Huo S, Yin Z, Tian Z, Huang F, Liu P, Liu Y, Yu F. Retracted and republished from: "The current state of research on influenza antiviral drug development: drugs in clinical trial and licensed drugs". mBio 2024; 15:e0017524. [PMID: 38551343 PMCID: PMC11077966 DOI: 10.1128/mbio.00175-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024] Open
Abstract
Influenza viruses (IVs) threaten global human health due to the high morbidity, infection, and mortality rates. Currently, the influenza drugs recommended by the Food and Drug Administration are oseltamivir, zanamivir, peramivir, and baloxavir marboxil. These recommended antivirals are currently effective for major subtypes of IVs as the compounds target conserved domains in neuraminidase or polymerase acidic (PA) protein. However, this trend may gradually change due to the selection of antiviral drugs and the natural evolution of IVs. Therefore, there is an urgent need to develop drugs related to the treatment of influenza to deal with the next pandemic. Here, we summarized the cutting-edge research in mechanism of action, inhibitory activity, and clinical efficacy of drugs that have been approved and drugs that are still in clinical trials for influenza treatment. We hope this review will provide up-to-date and comprehensive information on influenza antivirals and generate hypotheses for screens and development of new broad-spectrum influenza drugs in the near future.
Collapse
Affiliation(s)
- Yanbai Li
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zhe Yin
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zuguang Tian
- Department of High-Tech Development, Baoding City Science and Technology Bureau, Baoding, China
| | - Fang Huang
- Epidemic Prevention Laboratory, Tongzhou District Center For Animal Disease Control and Prevention, Beijing, China
| | - Peng Liu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yue Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| |
Collapse
|
5
|
Focosi D, Franchini M, Senefeld JW, Joyner MJ, Sullivan DJ, Pekosz A, Maggi F, Casadevall A. Passive immunotherapies for the next influenza pandemic. Rev Med Virol 2024; 34:e2533. [PMID: 38635404 DOI: 10.1002/rmv.2533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/20/2024]
Abstract
Influenzavirus is among the most relevant candidates for a next pandemic. We review here the phylogeny of former influenza pandemics, and discuss candidate lineages. After briefly reviewing the other existing antiviral options, we discuss in detail the evidences supporting the efficacy of passive immunotherapies against influenzavirus, with a focus on convalescent plasma.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Mantua Hospital, Mantua, Italy
| | - Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Fabrizio Maggi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Li Y, Huo S, Yin Z, Tian Z, Huang F, Liu P, Liu Y, Yu F. The current state of research on influenza antiviral drug development: drugs in clinical trial and licensed drugs. mBio 2023; 14:e0127323. [PMID: 37610204 PMCID: PMC10653855 DOI: 10.1128/mbio.01273-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Influenza viruses (IVs) threaten global human health due to the high morbidity, infection, and mortality rates. Currently, the influenza drugs recommended by the FDA are oseltamivir, zanamivir, peramivir, and baloxavir marboxil. Notably, owing to the high variability of IVs, no drug exists that can effectively treat all types and subtypes of IVs. Moreover, the current trend of drug resistance is likely to continue as the viral genome is constantly mutating. Therefore, there is an urgent need to develop drugs related to the treatment of influenza to deal with the next pandemic. Here, we summarized the cutting-edge research in mechanism of action, inhibitory activity, and clinical efficacy of drugs that have been approved and drugs that are still in clinical trials for influenza treatment. We hope this review will provide up-to-date and comprehensive information on influenza antivirals and generate hypotheses for screens and development of new broad-spectrum influenza drugs in the near future.
Collapse
Affiliation(s)
- Yanbai Li
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zhe Yin
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zuguang Tian
- Baoding City Science and Technology Bureau, Baoding, China
| | - Fang Huang
- Tongzhou District Center For Animal Disease Control and Prevention, Beijing, China
| | - Peng Liu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yue Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| |
Collapse
|
7
|
Gulland A. 'Gagged and blindsided': how an allegation of research misconduct affected our lab. Nature 2023:10.1038/d41586-023-02711-5. [PMID: 37626219 DOI: 10.1038/d41586-023-02711-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
|
8
|
Vanderven HA, Wentworth DN, Han WM, Peck H, Barr IG, Davey RT, Beigel JH, Dwyer DE, Jain MK, Angus B, Brandt CT, Mykietiuk A, Law MG, Neaton JD, Kent SJ. Understanding the treatment benefit of hyperimmune anti-influenza intravenous immunoglobulin (Flu-IVIG) for severe human influenza. JCI Insight 2023; 8:e167464. [PMID: 37289541 PMCID: PMC10443807 DOI: 10.1172/jci.insight.167464] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUNDAntibody-based therapies for respiratory viruses are of increasing importance. The INSIGHT 006 trial administered anti-influenza hyperimmune intravenous immunoglobulin (Flu-IVIG) to patients hospitalized with influenza. Flu-IVIG treatment improved outcomes in patients with influenza B but showed no benefit for influenza A.METHODSTo probe potential mechanisms of Flu-IVIG utility, sera collected from patients hospitalized with influenza A or B viruses (IAV or IBV) were analyzed for antibody isotype/subclass and Fcγ receptor (FcγR) binding by ELISA, bead-based multiplex, and NK cell activation assays.RESULTSInfluenza-specific FcγR-binding antibodies were elevated in Flu-IVIG-infused IBV- and IAV-infected patients. In IBV-infected participants (n = 62), increased IgG3 and FcγR binding were associated with more favorable outcomes. Flu-IVIG therapy also improved the odds of a more favorable outcome in patients with low levels of anti-IBV Fc-functional antibody. Higher FcγR-binding antibody was associated with less favorable outcomes in IAV-infected patients (n = 50), and Flu-IVIG worsened the odds of a favorable outcome in participants with low levels of anti-IAV Fc-functional antibody.CONCLUSIONThese detailed serological analyses provide insights into antibody features and mechanisms required for a successful humoral response against influenza, suggesting that IBV-specific, but not IAV-specific, antibodies with Fc-mediated functions may assist in improving influenza outcome. This work will inform development of improved influenza immunotherapies.TRIAL REGISTRATIONClinicalTrials.gov NCT02287467.FUNDINGFunding for this research was provided by subcontract 13XS134 under Leidos Biomedical Research Prime Contract HHSN261200800001E and HHSN261201500003I, NCI/NIAID.
Collapse
Affiliation(s)
- Hillary A. Vanderven
- Biomedicine, College of Public Health, Medical and Veterinary Sciences, and
- Australian Institute of Tropical Health and Medicine, James Cook University, Douglas, Queensland, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Deborah N. Wentworth
- Divison of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Win Min Han
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Heidi Peck
- WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Ian G. Barr
- WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Richard T. Davey
- National Institute of Allergy and Infectious Disease (NIAID), Bethesda, Maryland, USA
| | - John H. Beigel
- National Institute of Allergy and Infectious Disease (NIAID), Bethesda, Maryland, USA
| | - Dominic E. Dwyer
- New South Wales Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, Australia
| | | | - Brian Angus
- Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Christian T. Brandt
- Department of Infectious Diseases, Zealand University Hospital Roskilde, Denmark
| | | | - Matthew G. Law
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - James D. Neaton
- Divison of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stephen J. Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Carlton, Victoria, Australia
| | | |
Collapse
|
9
|
Struble EB, Rawson JMO, Stantchev T, Scott D, Shapiro MA. Uses and Challenges of Antiviral Polyclonal and Monoclonal Antibody Therapies. Pharmaceutics 2023; 15:pharmaceutics15051538. [PMID: 37242780 DOI: 10.3390/pharmaceutics15051538] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Viral diseases represent a major public health concerns and ever-present risks for developing into future pandemics. Antiviral antibody therapeutics, either alone or in combination with other therapies, emerged as valuable preventative and treatment options, including during global emergencies. Here we will discuss polyclonal and monoclonal antiviral antibody therapies, focusing on the unique biochemical and physiological properties that make them well-suited as therapeutic agents. We will describe the methods of antibody characterization and potency assessment throughout development, highlighting similarities and differences between polyclonal and monoclonal products as appropriate. In addition, we will consider the benefits and challenges of antiviral antibodies when used in combination with other antibodies or other types of antiviral therapeutics. Lastly, we will discuss novel approaches to the characterization and development of antiviral antibodies and identify areas that would benefit from additional research.
Collapse
Affiliation(s)
- Evi B Struble
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Jonathan M O Rawson
- Division of Antivirals, Office of Infectious Diseases, Office of New Drugs, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Tzanko Stantchev
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Dorothy Scott
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Office of Therapeutic Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Marjorie A Shapiro
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
10
|
Kumari R, Sharma SD, Kumar A, Ende Z, Mishina M, Wang Y, Falls Z, Samudrala R, Pohl J, Knight PR, Sambhara S. Antiviral Approaches against Influenza Virus. Clin Microbiol Rev 2023; 36:e0004022. [PMID: 36645300 PMCID: PMC10035319 DOI: 10.1128/cmr.00040-22] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Preventing and controlling influenza virus infection remains a global public health challenge, as it causes seasonal epidemics to unexpected pandemics. These infections are responsible for high morbidity, mortality, and substantial economic impact. Vaccines are the prophylaxis mainstay in the fight against influenza. However, vaccination fails to confer complete protection due to inadequate vaccination coverages, vaccine shortages, and mismatches with circulating strains. Antivirals represent an important prophylactic and therapeutic measure to reduce influenza-associated morbidity and mortality, particularly in high-risk populations. Here, we review current FDA-approved influenza antivirals with their mechanisms of action, and different viral- and host-directed influenza antiviral approaches, including immunomodulatory interventions in clinical development. Furthermore, we also illustrate the potential utility of machine learning in developing next-generation antivirals against influenza.
Collapse
Affiliation(s)
- Rashmi Kumari
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Department of Anesthesiology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Suresh D. Sharma
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Amrita Kumar
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Zachary Ende
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Oak Ridge Institute for Science and Education (ORISE), CDC Fellowship Program, Oak Ridge, Tennessee, USA
| | - Margarita Mishina
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Yuanyuan Wang
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Association of Public Health Laboratories, Silver Spring, Maryland, USA
| | - Zackary Falls
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Ram Samudrala
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Jan Pohl
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Paul R. Knight
- Department of Anesthesiology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Suryaprakash Sambhara
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
11
|
Switching Heavy Chain Constant Domains Denatures the Paratope 3D Architecture of Influenza Monoclonal Antibodies. Pathogens 2022; 12:pathogens12010051. [PMID: 36678399 PMCID: PMC9865026 DOI: 10.3390/pathogens12010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Several human monoclonal Abs for treating Influenza have been evaluated in clinical trials with limited success despite demonstrating superiority in preclinical animal models including mice. To conduct efficacy studies in mice, human monoclonal Abs are genetically engineered to contain mouse heavy chain constant domain to facilitate the engagement of Fc-receptors on mouse immune effector cells. Although studies have consistently reported discrepancies in Ab effectiveness following genetic engineering, the structural and mechanistic basis for these inconsistencies remain uncharacterized. Here, we use homology modeling to predict variable region (VR) analogous monoclonal Abs possessing human IgG1, mouse IgG1, and mouse IgG2a heavy chain constant domains. We then examine predicted 3D structures for variations in the spatial location and orientation of corresponding paratope amino acid residues. By structurally aligning crystal structures of Fabs in complex with hemagglutinin (HA), we show that corresponding paratope amino acid residues for VR-analogous human IgG1, mouse IgG1, and mouse IgG2a monoclonal Abs interact differentially with HA suggesting that their epitopes might not be identical. To demonstrate that variations in the paratope 3D fine architecture have implications for Ab specificity and effectiveness, we genetically engineered VR-analogous human IgG1, human IgG4, mouse IgG1, and mouse IgG2a monoclonal Abs and explored their specificity and effectiveness in protecting MDCK cells from infection by pandemic H1N1 and H3N2 Influenza viruses. We found that VR-analogous monoclonal Abs placed on mouse heavy chain constant domains were more efficacious at protecting MDCK cells from Influenza virus infection relative to those on human heavy chain constant domains. Interestingly, mouse but not human heavy chain constant domains increased target breadth in some monoclonal Abs. These data suggest that heavy chain constant domain sequences play a role in shaping Ab repertoires that go beyond class or sub-class differences in immune effector recruitment. This represents a facet of Ab biology that can potentially be exploited to improve the scope and utilization of current therapeutic or prophylactic candidates for influenza.
Collapse
|
12
|
Broad-Spectrum Activity of Small Molecules Acting against Influenza a Virus: Biological and Computational Studies. Pharmaceuticals (Basel) 2022; 15:ph15030301. [PMID: 35337099 PMCID: PMC8952214 DOI: 10.3390/ph15030301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 01/25/2023] Open
Abstract
Influenza still represents a problematic disease, involving millions of people every year and causing hundreds of thousands of deaths. Only a few drugs are clinically available. The search for an effective weapon is still ongoing. In this scenario, we recently identified new drug-like compounds with antiviral activity toward two A/H1N1 Influenza virus strains, which were demonstrated to interfere with the processes mediated by hemagglutinin (HA). In the present work, the compound’s ability to act against the A/H3N2 viral strain has been evaluated in hemagglutination inhibition (HI) assays. Two of the five tested compounds were also active toward the A/H3N2 Influenza virus. To validate the scaffold activity, analogue compounds of two broad-spectrum molecules were selected and purchased for HI testing on both A/H1N1 and A/H3N2 Influenza viruses. Forty-three compounds were tested, and four proved to be active toward all three viral strains. A computational study has been carried out to depict the HA binding process of the most interesting compounds.
Collapse
|
13
|
A broadly neutralizing human monoclonal antibody against the hemagglutinin of avian influenza virus H7N9. Chin Med J (Engl) 2022; 135:799-805. [PMID: 35671181 PMCID: PMC9276153 DOI: 10.1097/cm9.0000000000002001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: The new emerging avian influenza A H7N9 virus, causing severe human infection with a mortality rate of around 41%. This study aims to provide a novel treatment option for the prevention and control of H7N9. Methods: H7 hemagglutinin (HA)-specific B cells were isolated from peripheral blood plasma cells of the patients previously infected by H7N9 in Jiangsu Province, China. The human monoclonal antibodies (mAbs) were generated by amplification and cloning of these HA-specific B cells. First, all human mAbs were screened for binding activity by enzyme-linked immunosorbent assay. Then, those mAbs, exhibiting potent affinity to recognize H7 HAs were further evaluated by hemagglutination-inhibiting (HAI) and microneutralization in vitro assays. Finally, the lead mAb candidate was selected and tested against the lethal challenge of the H7N9 virus using murine models. Results: The mAb 6-137 was able to recognize a panel of H7 HAs with high affinity but not HA of other subtypes, including H1N1 and H3N2. The mAb 6-137 can efficiently inhibit the HA activity in the inactivated H7N9 virus and neutralize 100 tissue culture infectious dose 50 (TCID50) of H7N9 virus (influenza A/Nanjing/1/2013) in vitro, with neutralizing activity as low as 78 ng/mL. In addition, the mAb 6-137 protected the mice against the lethal challenge of H7N9 prophylactically and therapeutically. Conclusion: The mAb 6-137 could be an effective antibody as a prophylactic or therapeutic biological treatment for the H7N9 exposure or infection.
Collapse
|
14
|
Voronina DV, Shcheblyakov DV, Esmagambetov IB, Derkaev AA, Popova O, Shcherbinin DN. Development of Neutralizing Nanobodies to the Hemagglutinin Stem Domain of Influenza A Viruses. Acta Naturae 2021; 13:33-41. [PMID: 35127144 PMCID: PMC8807538 DOI: 10.32607/actanaturae.11495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/05/2021] [Indexed: 11/20/2022] Open
Abstract
The influenza virus infection claims ~650,000 lives annually. Taking into account the evolving resistance of the pathogen to antiviral drugs and the waning effectiveness of vaccination among certain populations, new approaches to the treatment of influenza are needed. The current study is aimed at obtaining single-domain antibodies (Nanobodies®) to the highly conserved stem domain of influenza A virus hemagglutinin by phage display. Two high-affinity neutralizing clones of Nanobodies® with a particular specificity were selected; they ensured 100% neutralization of the H1N1 and H5N2 influenza viruses in vivo. The obtained data demonstrate that it is possible to develop highly effective VHH-based drugs for the treatment of influenza.
Collapse
Affiliation(s)
- D. V. Voronina
- FSBI “National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of Russia, Moscow, 123098 Russia
| | - D. V. Shcheblyakov
- FSBI “National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of Russia, Moscow, 123098 Russia
| | - I. B. Esmagambetov
- FSBI “National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of Russia, Moscow, 123098 Russia
| | - A. A. Derkaev
- FSBI “National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of Russia, Moscow, 123098 Russia
| | - O. Popova
- FSBI “National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of Russia, Moscow, 123098 Russia
| | - D. N. Shcherbinin
- FSBI “National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of Russia, Moscow, 123098 Russia
| |
Collapse
|
15
|
Yu Y, Wang M, Zhang X, Li S, Lu Q, Zeng H, Hou H, Li H, Zhang M, Jiang F, Wu J, Ding R, Zhou Z, Liu M, Si W, Zhu T, Li H, Ma J, Gu Y, She G, Li X, Zhang Y, Peng K, Huang W, Liu W, Wang Y. Antibody-dependent cellular cytotoxicity response to SARS-CoV-2 in COVID-19 patients. Signal Transduct Target Ther 2021; 6:346. [PMID: 34561414 PMCID: PMC8463587 DOI: 10.1038/s41392-021-00759-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) responses to viral infection are a form of antibody regulated immune responses mediated through the Fc fragment. Whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered ADCC responses contributes to COVID-19 disease development is currently not well understood. To understand the potential correlation between ADCC responses and COVID-19 disease development, we analyzed the ADCC activity and neutralizing antibody response in 255 individuals ranging from asymptomatic to fatal infections over 1 year post disease. ADCC was elicited by 10 days post-infection, peaked by 11-20 days, and remained detectable until 400 days post-infection. In general, patients with severe disease had higher ADCC activities. Notably, patients who had severe disease and recovered had higher ADCC activities than patients who had severe disease and deceased. Importantly, ADCC activities were mediated by a diversity of epitopes in SARS-COV-2-infected mice and induced to comparable levels against SARS-CoV-2 variants of concern (VOCs) (B.1.1.7, B.1.351, and P.1) as that against the D614G mutant in human patients and vaccinated mice. Our study indicates anti-SARS-CoV-2 ADCC as a major trait of COVID-19 patients with various conditions, which can be applied to estimate the extra-neutralization level against COVID-19, especially lethal COVID-19.
Collapse
Affiliation(s)
- Yuanling Yu
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Meiyu Wang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China ,grid.506261.60000 0001 0706 7839Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoai Zhang
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Shufen Li
- grid.9227.e0000000119573309State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei China
| | - Qingbin Lu
- grid.11135.370000 0001 2256 9319Department of Laboratorial Science and Technology, School of Public Health, Peking University, Beijing, China
| | - Haolong Zeng
- grid.33199.310000 0004 0368 7223Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Hou
- grid.33199.310000 0004 0368 7223Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Li
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Mengyi Zhang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Fei Jiang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Jiajing Wu
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Ruxia Ding
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Zehua Zhou
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Min Liu
- grid.274690.eSinovac Biotech Co., Ltd, Beijing, China
| | - Weixue Si
- Cansino Biotech Incorporation, Tianjin, China
| | - Tao Zhu
- Cansino Biotech Incorporation, Tianjin, China
| | - Hangwen Li
- Stemirna Therapeutics, Ltd, Shanghai, China
| | - Jie Ma
- Stemirna Therapeutics, Ltd, Shanghai, China
| | | | - Guangbiao She
- Anhui Zhifeilongcom Biopharmaceutical Co., Ltd, Hefei, China
| | - Xiaokun Li
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yulan Zhang
- grid.9227.e0000000119573309State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei China
| | - Ke Peng
- grid.9227.e0000000119573309State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Weijin Huang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Wei Liu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Youchun Wang
- grid.410749.f0000 0004 0577 6238Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China ,grid.506261.60000 0001 0706 7839Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Beigel JH, Hayden FG. Influenza Therapeutics in Clinical Practice-Challenges and Recent Advances. Cold Spring Harb Perspect Med 2021; 11:a038463. [PMID: 32041763 PMCID: PMC8015700 DOI: 10.1101/cshperspect.a038463] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the last few years, several new direct-acting influenza antivirals have been licensed, and others have advanced in clinical development. The increasing diversity of antiviral classes should allow an adequate public health response should a resistant virus to one agent or class widely circulate. One new antiviral, baloxavir marboxil, has been approved in the United States for treatment of influenza in those at high risk of developing influenza-related complications. Except for intravenous zanamivir in European Union countries, no antivirals have been licensed specifically for the indication of severe influenza or hospitalized influenza. This review addresses recent clinical developments involving selected polymerase inhibitors, neuraminidase inhibitors, antibody-based therapeutics, and host-directed therapies. There are many knowledge gaps for most of these agents because some data are not published and multiple pivotal studies are in progress at present. This review also considers important clinical research issues, including regulatory pathways, study designs, endpoints, and target populations encountered during the clinical development of novel therapeutics.
Collapse
Affiliation(s)
- John H Beigel
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20892-9826, USA
| | - Frederick G Hayden
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| |
Collapse
|
17
|
Chiang YW, Li CJ, Su HY, Hsieh KT, Weng CW, Chen HW, Chang SC. Development of mouse monoclonal antibody for detecting hemagglutinin of avian influenza A(H7N9) virus and preventing virus infection. Appl Microbiol Biotechnol 2021; 105:3235-3248. [PMID: 33770244 PMCID: PMC7995400 DOI: 10.1007/s00253-021-11253-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/15/2021] [Accepted: 03/21/2021] [Indexed: 12/29/2022]
Abstract
Abstract Many cases of avian influenza A(H7N9) virus infection in humans have been reported since its first emergence in 2013. The disease is of concern because most patients have become severely ill with roughly 30% mortality rate. Because the threat in public health caused by H7N9 virus remains high, advance preparedness is essentially needed. In this study, the recombinant H7N9 hemagglutinin (HA) was expressed in insect cells and purified for generation of two monoclonal antibodies, named F3-2 and 1C6B. F3-2 can only recognize the H7N9 HA without having cross-reactivity with HA proteins of H1N1, H3N2, H5N1, and H7N7. 1C6B has the similar specificity with F3-2, but 1C6B can also bind to H7N7 HA. The binding epitope of F3-2 is mainly located in the region of H7N9 HA(299–307). The binding epitope of 1C6B is located in the region of H7N9 HA(489–506). F3-2 and 1C6B could not effectively inhibit the hemagglutination activity of H7N9 HA. However, F3-2 can prevent H7N9 HA from trypsin cleavage and can bind to H7N9 HA which has undergone pH-induced conformational change. F3-2 also has the ability of binding to H7N9 viral particles and inhibiting H7N9 virus infection to MDCK cells with the IC50 value of 22.18 μg/mL. In addition, F3-2 and 1C6B were utilized for comprising a lateral flow immunochromatographic test strip for specific detection of H7N9 HA. Key points • Two mouse monoclonal antibodies, F3-2 and 1C6B, were generated for recognizing the novel binding epitopes in H7N9 HA. • F3-2 can prevent H7N9 HA from trypsin cleavage and inhibit H7N9 virus infection to MDCK cells. • F3-2 and 1C6B were developed as a lateral flow immunochromatographic test for specific detection of H7N9 HA.
Collapse
Affiliation(s)
- Yi-Wei Chiang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Chia-Jung Li
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Heng-Yi Su
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Kai-Ting Hsieh
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Chia-Wei Weng
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan
| | - Shih-Chung Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 106, Taiwan.
- Center of Biotechnology, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
18
|
Zhou J, Krishnan N, Jiang Y, Fang RH, Zhang L. Nanotechnology for virus treatment. NANO TODAY 2021; 36:101031. [PMID: 33519948 PMCID: PMC7836394 DOI: 10.1016/j.nantod.2020.101031] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 04/14/2023]
Abstract
The continued emergence of novel viruses poses a significant threat to global health. Uncontrolled outbreaks can result in pandemics that have the potential to overburden our healthcare and economic systems. While vaccination is a conventional modality that can be employed to promote herd immunity, antiviral vaccines can only be applied prophylactically and do little to help patients who have already contracted viral infections. During the early stages of a disease outbreak when vaccines are unavailable, therapeutic antiviral drugs can be used as a stopgap solution. However, these treatments do not always work against emerging viral strains and can be accompanied by adverse effects that sometimes outweigh the benefits. Nanotechnology has the potential to overcome many of the challenges facing current antiviral therapies. For example, nanodelivery vehicles can be employed to drastically improve the pharmacokinetic profile of antiviral drugs while reducing their systemic toxicity. Other unique nanomaterials can be leveraged for their virucidal or virus-neutralizing properties. In this review, we discuss recent developments in antiviral nanotherapeutics and provide a perspective on the application of nanotechnology to the SARS-CoV-2 outbreak and future virus pandemics.
Collapse
Affiliation(s)
- Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Jiang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
19
|
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: 4.0] [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
|
20
|
Huang J, Huang N, Fan M, Zhao L, Luo Y, Ding P, Tian M, Liu Q, Guo Y, Zhao J, Zheng Y, Zhang H, Ping J. Hemagglutinin stalk-based monoclonal antibody elicits broadly reactivity against group 1 influenza A virus. Virol J 2020; 17:191. [PMID: 33287849 PMCID: PMC7720065 DOI: 10.1186/s12985-020-01458-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/23/2020] [Indexed: 11/23/2022] Open
Abstract
Background Influenza virus remains a continuous and severe threat to public health worldwide, and its prevention and treatment have always been a major international issue. Because of its ability to evade immune surveillance through rapid antigenic drift and antigenic shift, broad-spectrum vaccines seem increasingly important. Methods A mAb named 3C12 from an immortalized hybrid cell was generated via immunizing mice with HA2 protein from A/chicken/Anhui/BRI99/2016 (AH/BRI99/16, H9N2) generated by prokaryotic expression. Then, its broad-spectrum activity was analyzed by WB and IFA. Next, the minimal linear epitope was identified via analyzing the reaction of a series of HA truncations with 3C12. Finally, the protective effects of 3C12 were evaluated in vitro and in vivo infection experiments.
Results The mAb could react with the viruses of subtypes H1, H2, H5, H8, H9, H12, H13, H16, and HA protein of H18 in group 1, but failed to react with viruses in group 2. The minimal linear epitope targeted by the mAb was 433NAELLVL439 in full length of HA and localized in the C-helix region of HA2 (residue 95-101, HA2 numbering). What’s more, the mAb 3C12 inhibited H1, H2, H5, H8, H9, H12, H13 and H16 virus-replication in vitro and also has shown effectiveness in preventing and treating disease in mice challenged with lethal dose of AH/BRI99/16 (H9N2) virus in vivo. These results suggested that the broadly reactive anti-HA stem mAb 3C12 exhibited prophylactic and therapeutic efficacy. Conclusions Here, we have demonstrated that the linear epitope identified in this study could be a novel target for developing broad-spectrum influenza diagnostics or vaccine design, and the HA2-based monoclonal antibody is indeed a promising strategy for broad-spectrum protection against seasonal and pandemic influenza viruses.
Collapse
Affiliation(s)
- Jingjin Huang
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Nan Huang
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Menglu Fan
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lingcai Zhao
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yan Luo
- Biotechnology Research laboratory, Jiangsu Lihua Animal Husbandry co. LTD, Changzhou, 213168, China
| | - Pingyun Ding
- Biotechnology Research laboratory, Jiangsu Lihua Animal Husbandry co. LTD, Changzhou, 213168, China
| | - Miao Tian
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qingzheng Liu
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yanna Guo
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jinhua Zhao
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yiqing Zheng
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Haitao Zhang
- Biotechnology Research laboratory, Jiangsu Lihua Animal Husbandry co. LTD, Changzhou, 213168, China.
| | - Jihui Ping
- MOE International Joint Collaborative Research Laboratory for Animal Health and Food Safety & Jiangsu Engineering Laboratory of Animal Immunology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
21
|
Heida R, Bhide YC, Gasbarri M, Kocabiyik Ö, Stellacci F, Huckriede ALW, Hinrichs WLJ, Frijlink HW. Advances in the development of entry inhibitors for sialic-acid-targeting viruses. Drug Discov Today 2020; 26:122-137. [PMID: 33099021 PMCID: PMC7577316 DOI: 10.1016/j.drudis.2020.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/13/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
Over the past decades, several antiviral drugs have been developed to treat a range of infections. Yet the number of treatable viral infections is still limited, and resistance to current drug regimens is an ever-growing problem. Therefore, additional strategies are needed to provide a rapid cure for infected individuals. An interesting target for antiviral drugs is the process of viral attachment and entry into the cell. Although most viruses use distinct host receptors for attachment to the target cell, some viruses share receptors, of which sialic acids are a common example. This review aims to give an update on entry inhibitors for a range of sialic-acid-targeting viruses and provides insight into the prospects for those with broad-spectrum potential.
Collapse
Affiliation(s)
- Rick Heida
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| | - Yoshita C Bhide
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands; Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713AV Groningen, The Netherlands
| | - Matteo Gasbarri
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Özgün Kocabiyik
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Anke L W Huckriede
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713AV Groningen, The Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands.
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| |
Collapse
|
22
|
Broader neutralization of CT-P27 against influenza A subtypes by combining two human monoclonal antibodies. PLoS One 2020; 15:e0236172. [PMID: 32726321 PMCID: PMC7390384 DOI: 10.1371/journal.pone.0236172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/30/2020] [Indexed: 12/02/2022] Open
Abstract
There are several broadly neutralizing monoclonal antibodies that neutralize influenza viruses with different mechanisms from traditional polyclonal antibodies induced by vaccination. CT149, which is one of the broadly neutralizing antibodies, was also previously reported to neutralize group 2 and some of group 1 influenza viruses (13 out of 13 tested group 2 viruses and 5 out of 11 group 1 viruses). In this study, we developed another antibody with the aim of compensating partial coverage of CT149 against group 1 influenza viruses. CT120 was screened among different antibody candidates and mixed with CT149. Importantly, although the binding sites of CT120 and CT149 are close to each other, the two antibodies do not interfere. The mixture of CT120 and CT149, which we named as CT-P27, showed broad efficacy by neutralizing 37 viruses from 11 different subtypes, of both group 1 and 2 influenza A viruses. Moreover, CT-P27 showed in vivo therapeutic efficacy, long prophylactic potency, and synergistic effect with oseltamivir in influenza virus-challenged mouse models. Our findings provide a novel therapeutic opportunity for more efficient treatment of influenza.
Collapse
|
23
|
Li Y, Wang L, Si H, Yu Z, Tian S, Xiang R, Deng X, Liang R, Jiang S, Yu F. Influenza virus glycoprotein-reactive human monoclonal antibodies. Microbes Infect 2020; 22:263-271. [PMID: 32569735 PMCID: PMC7303604 DOI: 10.1016/j.micinf.2020.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/08/2020] [Indexed: 11/05/2022]
Abstract
Influenza continues to be a significant public health challenge. Two glycoproteins on the surface of influenza virus, hemagglutinin and neuraminidase, play a prominent role in the process of influenza virus infection and release. Monoclonal antibodies targeting glycoproteins can effectively prevent the spread of the virus. In this review, we summarized currently reported human monoclonal antibodies targeting glycoproteins of influenza A and B viruses.
Collapse
Affiliation(s)
- Yanbai Li
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Lili Wang
- Research Center of Chinese Jujube, Hebei Agricultural University, Baoding, China
| | - Helong Si
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zhengsen Yu
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shijun Tian
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Rong Xiang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Xiaoqian Deng
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Ruiying Liang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Fei Yu
- College of Life Sciences, Hebei Agricultural University, Baoding, China.
| |
Collapse
|
24
|
De Jong NMC, Aartse A, Van Gils MJ, Eggink D. Development of broadly reactive influenza vaccines by targeting the conserved regions of the hemagglutinin stem and head domains. Expert Rev Vaccines 2020; 19:563-577. [PMID: 32510256 DOI: 10.1080/14760584.2020.1777861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Influenza virus infections cause serious illness in millions of people each year. Although influenza virus vaccines are available, they are not optimally effective due to mismatches between the influenza virus strains used for the vaccine and the circulating strains. To improve protection by vaccines, a broadly protective or universal vaccine may be required. Strategies to develop universal vaccines aim to elicit broadly reactive antibodies, which target regions on the viral hemagglutinin (HA) protein which are conserved between strains. Broadly reactive antibodies have helped to identify such targets and can guide the design of such a vaccine. AREAS COVERED The first part of this review provides an in-depth overview of broadly reactive anti-HA antibodies, discussing their origin, breadth and their mechanisms of protection. The second part discusses the technical design and mode of action of potential universal vaccine candidates that aim to elicit these broadly reactive antibodies and provide protection against a majority of influenza strains. EXPERT OPINION While great strides have been made in the development of universal influenza vaccine candidates, real-life use still requires improvement of stability, enhancement of their breadth of protection and ease of production, while efficacies need to be determined in human trials.
Collapse
Affiliation(s)
- Nina M C De Jong
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam , Amsterdam, The Netherlands
| | - Aafke Aartse
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam , Amsterdam, The Netherlands.,Department of Virology, Biomedical Primate Research Centre , Rijswijk, The Netherlands
| | - Marit J Van Gils
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam , Amsterdam, The Netherlands
| | - Dirk Eggink
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam , Amsterdam, The Netherlands
| |
Collapse
|
25
|
Sloan SE, Szretter KJ, Sundaresh B, Narayan KM, Smith PF, Skurnik D, Bedard S, Trevejo JM, Oldach D, Shriver Z. Clinical and virological responses to a broad-spectrum human monoclonal antibody in an influenza virus challenge study. Antiviral Res 2020; 184:104763. [PMID: 32151645 DOI: 10.1016/j.antiviral.2020.104763] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/06/2020] [Accepted: 03/04/2020] [Indexed: 01/03/2023]
Abstract
Influenza A infections cause significant seasonal morbidity and mortality as well as periodic pandemic infections. Currently, no approved therapies exist for patients hospitalized with influenza. The efficacy of VIS410, a broadly neutralizing human immunoglobulin IgG1 monoclonal antibody engineered to bind to the stem region of group 1 and 2 influenza A hemagglutinins, was explored in experimental human influenza infection. Healthy volunteers were inoculated with influenza A/California/07/2009 (H1N1) and received a single dose of VIS410 or placebo 24 h later. Subjects were monitored for symptoms, viral shedding, and safety, including cytokine measurements. The primary efficacy endpoint was the area under the curve (AUC) of viral load (VL) in the VIS410 group versus placebo. VIS410 treatment was associated with a 76% reduction in median VL AUC as measured by qRT-PCR (p = 0.024). Similar VIS410 antiviral activity was observed by virus culture, with a 91% reduction in median VL AUC by TCID50 (p = 0.019) compared to placebo-treated volunteers. Influenza symptoms were generally mild or moderate, with a trend toward faster resolution in VIS410-treated subjects. Treatment with VIS410 was generally safe, with an increase in gastrointestinal events that were largely mitigated by pre-treatment with oral diphenhydramine (50 mg) in combination with 600 mg of ibuprofen. Transient elevation of specific cytokines (IL-8 and TNFα) were associated with gastrointestinal adverse events. Treatment with VIS410 did not interfere with the endogenous immune response to influenza A. These data indicate that VIS410 may provide therapeutic benefit in influenza A infection. TRIAL REGISTRATION: ClinicaTtrials.gov Identification NCT02468115; https://clinicaltrials.gov/ct2/show/NCT02468115?term=NCT02468115&rank=1).
Collapse
Affiliation(s)
| | - Kristy J Szretter
- Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts 02139, USA
| | | | | | | | - David Skurnik
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Institut Necker-Enfants Malades, INSERM U1151, CNRS UMR, 8253, Paris, France; Université Paris Descartes, Paris, France; Service de Microbiologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | | | | | | |
Collapse
|
26
|
Vogel OA, Manicassamy B. Broadly Protective Strategies Against Influenza Viruses: Universal Vaccines and Therapeutics. Front Microbiol 2020; 11:135. [PMID: 32117155 PMCID: PMC7020694 DOI: 10.3389/fmicb.2020.00135] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/21/2020] [Indexed: 12/21/2022] Open
Abstract
Influenza virus is a respiratory pathogen that can cause disease in humans, with symptoms ranging from mild to life-threatening. The vast majority of influenza virus infections in humans are observed during seasonal epidemics and occasional pandemics. Given the substantial public health burden associated with influenza virus infection, yearly vaccination is recommended for protection against seasonal influenza viruses. Despite vigilant surveillance for new variants and careful selection of seasonal vaccine strains, the efficacy of seasonal vaccines can vary widely from year to year. This often results in lowered protection within the population, regardless of vaccination status. In order to broaden the protection afforded by seasonal influenza vaccines, the National Institute of Allergy and Infectious Diseases (NIAID) has deemed the development of a universal influenza virus vaccine to be a priority in influenza virus vaccine research. This universal vaccine would provide protection against all influenza virus strains, eliminating the need for the yearly reformulations of seasonal influenza vaccines. In addition to universal influenza vaccine efforts, substantial progress has been made in developing novel influenza virus therapeutics that utilize broadly neutralizing antibodies to provide protection against influenza virus infection and to mitigate disease outcomes during infection. In this review, we discuss various approaches toward the goal of improving influenza virus vaccine efficacy through a universal influenza virus vaccine. We also address the novel methods of discovery and utilization of broadly neutralizing antibodies to improve influenza disease outcomes.
Collapse
Affiliation(s)
- Olivia A Vogel
- Department of Microbiology, The University of Chicago, Chicago, IL, United States
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
27
|
Mathew NR, Angeletti D. Recombinant Influenza Vaccines: Saviors to Overcome Immunodominance. Front Immunol 2020; 10:2997. [PMID: 31998299 PMCID: PMC6966699 DOI: 10.3389/fimmu.2019.02997] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/06/2019] [Indexed: 11/24/2022] Open
Abstract
It has been almost a decade since the 2009 influenza A virus pandemic hit the globe causing significant morbidity and mortality. Nonetheless, annual influenza vaccination, which elicits antibodies mainly against the head region of influenza hemagglutinin (HA), remains as the mainstay to combat and reduce symptoms of influenza infection. Influenza HA is highly antigenically variable, thus limiting vaccine efficacy. In addition, the variable HA head occupies the upper strata of the immunodominance hierarchy, thereby clouding the antibody response toward subdominant epitopes, which are usually conserved across different influenza strains. Isolation of monoclonal antibodies from individuals recognizing such epitopes has facilitated the development of recombinant vaccines that focus the adaptive immune response toward conserved, protective targets. Here, we review some significant leaps in recombinant vaccine development, which could possibly help to overcome B cell and antibody immunodominance and provide heterosubtypic immunity to influenza A virus.
Collapse
Affiliation(s)
- Nimitha R Mathew
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Davide Angeletti
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
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: 3.0] [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
|
29
|
Szollosi D, Bill A. Potential Role of Endonuclease Inhibition and Other Targets in the Treatment of Influenza. Curr Drug Targets 2019; 21:202-211. [PMID: 31368872 DOI: 10.2174/1389450120666190801115130] [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: 05/17/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Influenza is a single-stranded RNA virus that is highly contagious and infects millions of people in the U.S. annually. Due to complications, approximately 959,000 people were hospitalized and another 79,400 people died during the 2017-2018 flu season. While the best methods of prevention continue to be vaccination and hygiene, antiviral treatments may help reduce symptoms for those who are infected. Until recently, the only antiviral drugs in use have been the neuraminidase inhibitors: oseltamivir, zanamivir, and peramivir. OBJECTIVE We reviewed novel drug targets that can be used in the treatment of influenza, particularly in the case of neuraminidase inhibitor-resistant strains that may emerge. RESULTS More recently, a drug with a new mechanism of action has been approved. Baloxavir marboxil inhibits the influenza cap-dependent endonuclease that is needed for the virus to initiate replication within the host cell. This endonuclease target is within the polymerase acid (PA) subunit of RNA polymerase. Since the RNA-dependent RNA polymerase consists of two other subunits, polymerase basic 1 and 2, RNA polymerase has several targets that prevent viral replication. Other targets still under investigation include viral kinases, endocytosis, and viral fusion. CONCLUSION Due to the possibility of viral mutations and resistance, it is important to have antivirals with different mechanisms available, especially in the case of a new pandemic strain. Several novel antivirals are within various stages of development and may represent new classes of treatments that can reduce symptoms and complications in those patients who may be at higher risk.
Collapse
Affiliation(s)
- Doreen Szollosi
- University of Saint Joseph, School of Pharmacy & Physician Assistant Studies 229 Trumbull Street, Hartford, CT 06103, United States
| | - Ashley Bill
- University of Saint Joseph, School of Pharmacy & Physician Assistant Studies 229 Trumbull Street, Hartford, CT 06103, United States
| |
Collapse
|
30
|
Rapid isolation of a potent human antibody against H7N9 influenza virus from an infected patient. Antiviral Res 2019; 170:104564. [PMID: 31336147 DOI: 10.1016/j.antiviral.2019.104564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 11/23/2022]
Abstract
Influenza virus A H7N9 remains a serious threat to public health due to the lack of effective vaccines and drugs. In this study, a neutralizing human antibody named 3L11 was rapidly isolated from the switched memory B cells of a patient infected with H7N9. The antibody 3L11 was encoded by the heavy-chain VH1-8 gene and the light-chain VL2-13 gene that had undergone somatic mutations, and conferred high affinity binding to H7N9 hemagglutinins (HAs). It promoted killing of infected cells by antibody-dependent cell-mediated cytotoxicity (ADCC). Epitope mapping by mass spectroscopy (MS) indicated that 3L11 bound to the peptide 149-175 of HAs that contained the 150-loop of the receptor-binding site (RBS). Additionally, the 3L11 escape strains had G151R (Gly151→Arg151) and S152P (Ser152→Pro152) mutations within a conserved antigenic site A near the RBS that were not observed in field strains. Importantly, 3L11 fully protected mice against a lethal H7N9 virus challenge, in both pre- and postexposure administration regimens. Altogether, this work demonstrates the feasibility of rapid isolation of neutralizing H7N9 antibodies from infected patients and provides a potential prophylactic and therapeutic agent against H7N9 viruses.
Collapse
|
31
|
Sedeyn K, Saelens X. New antibody-based prevention and treatment options for influenza. Antiviral Res 2019; 170:104562. [PMID: 31323236 DOI: 10.1016/j.antiviral.2019.104562] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/16/2019] [Indexed: 12/23/2022]
Abstract
The antigenic diversity of human influenza viruses represents a challenge to the development of vaccines with durable immune protection. In addition, small molecule anti-influenza viral drugs can bring clinical relief to influenza patients but the emergence of drug resistant viruses can rapidly limit the effectiveness of such drugs. In the past decade, a number of human monoclonal antibodies have been described that can bind to and neutralize a broad range of influenza A and B viruses. Most of these monoclonal antibodies are directed against the viral hemagglutinin (HA) stalk and some have now been evaluated in early to mid-stage clinical trials. An important conclusion from these clinical studies is that hemagglutinin stalk-specific antibodies are safe and can reduce influenza symptoms. In addition, examples of bi- and multi-specific anti-influenza antibodies are discussed, although such antibodies have not yet progressed into clinical testing. In the future, antibody-based therapies might become part of our arsenal to prevent and treat influenza.
Collapse
Affiliation(s)
- Koen Sedeyn
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biochemistry and Microbiology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium; Department of Biochemistry and Microbiology, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
| |
Collapse
|
32
|
Finlay WJ, Lugovskoy AA. De novo discovery of antibody drugs - great promise demands scrutiny. MAbs 2019; 11:809-811. [PMID: 31122133 PMCID: PMC6601558 DOI: 10.1080/19420862.2019.1622926] [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: 05/19/2019] [Revised: 05/01/2019] [Accepted: 05/21/2019] [Indexed: 01/10/2023] Open
Abstract
We live in an era of rapidly advancing computing capacity and algorithmic sophistication. "Big data" and "artificial intelligence"find progressively wider use in all spheres of human activity, including healthcare. A diverse array of computational technologies is being applied with increasing frequency to antibody drug research and development (R&D). Their successful applications are met with great interest due to the potential for accelerating and streamlining the antibody R&D process. While this excitement is very likely justified in the long term, it is less likely that the transition from the first use to routine practice will escape challenges that other new technologies had experienced before they began to blossom. This transition typically requires many cycles of iterative learning that rely on the deconstruction of the technology to understand its pitfalls and define vectors for optimization. The study by Vasquez et al. identifies a key obstacle to such learning: the lack of transparency regarding methodology in computational antibody design reports, which has the potential to mislead the community efforts.
Collapse
|
33
|
Beigel JH, Nam HH, Adams PL, Krafft A, Ince WL, El-Kamary SS, Sims AC. Advances in respiratory virus therapeutics - A meeting report from the 6th isirv Antiviral Group conference. Antiviral Res 2019; 167:45-67. [PMID: 30974127 PMCID: PMC7132446 DOI: 10.1016/j.antiviral.2019.04.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/11/2022]
Abstract
The International Society for Influenza and other Respiratory Virus Diseases held its 6th Antiviral Group (isirv-AVG) conference in Rockville, Maryland, November 13-15, 2018. The three-day program was focused on therapeutics towards seasonal and pandemic influenza, respiratory syncytial virus, coronaviruses including MERS-CoV and SARS-CoV, human rhinovirus, and other respiratory viruses. Updates were presented on several influenza antivirals including baloxavir, CC-42344, VIS410, immunoglobulin, immune plasma, MHAA4549A, pimodivir (JNJ-63623872), umifenovir, and HA minibinders; RSV antivirals including presatovir (GS-5806), ziresovir (AK0529), lumicitabine (ALS-008176), JNJ-53718678, JNJ-64417184, and EDP-938; broad spectrum antivirals such as favipiravir, VH244, remdesivir, and EIDD-1931/EIDD-2801; and host directed strategies including nitazoxanide, eritoran, and diltiazem. Other topics included considerations of novel endpoints such as ordinal scales and patient reported outcomes (PRO), and study design issues, and other regulatory considerations for antiviral drug development. The aim of this report is to provide a summary of the presentations given at this meeting.
Collapse
Affiliation(s)
- John H Beigel
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Hannah H Nam
- (b)Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Peter L Adams
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS), Washington, DC, USA
| | - Amy Krafft
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - William L Ince
- Division of Antiviral Products, Office of Antimicrobial Products, Office of New Drugs, Center for Drug Evaluation and Research, U.S Food and Drug Administration, Silver Spring, MD, USA
| | - Samer S El-Kamary
- Division of Antiviral Products, Office of Antimicrobial Products, Office of New Drugs, Center for Drug Evaluation and Research, U.S Food and Drug Administration, Silver Spring, MD, USA
| | - Amy C Sims
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Vásquez M, Krauland E, Walker L, Wittrup D, Gerngross T. Connecting the sequence dots: shedding light on the genesis of antibodies reported to be designed in silico. MAbs 2019; 11:803-808. [PMID: 31107637 PMCID: PMC6601546 DOI: 10.1080/19420862.2019.1611172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Two recent publications out of the same research laboratory report on structure-based in silico design of antibodies against viral targets without sequence disclosure. Cross-referencing the published data to patent databases, we established the sequence identity of said computationally designed antibodies. In both cases, the antibodies align with high sequence identity to previously reported antibodies of the same specificity. This clear underlying sequence relationship, which is far closer than the antibody templates reported to seed the computational design, suggests an alternative origin of the computationally designed antibodies. The lack of both reproducible computational algorithms and of output sequences in the initial publications obscures the relationship to previously reported antibodies, and sows doubt as to the genesis narrative described therein.
Collapse
Affiliation(s)
| | | | | | - Dane Wittrup
- a Adimab LLC , Lebanon , NH , USA.,b Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Tillman Gerngross
- a Adimab LLC , Lebanon , NH , USA.,c Thayer School of Engineering , Dartmouth College , Hanover , NH , USA
| |
Collapse
|
35
|
Xiao H, Guo T, Yang M, Qi J, Huang C, Hong Y, Gu J, Pang X, Liu WJ, Peng R, McCauley J, Bi Y, Li S, Feng J, Zhang H, Zhang X, Lu X, Yan J, Chen L, Shi Y, Chen W, Gao GF. Light chain modulates heavy chain conformation to change protection profile of monoclonal antibodies against influenza A viruses. Cell Discov 2019; 5:21. [PMID: 30993000 PMCID: PMC6465249 DOI: 10.1038/s41421-019-0086-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
The isolation of human monoclonal antibodies with broadly neutralizing breadth can provide a promising countermeasure for influenza A viruses infection. Most broadly neutralizing antibodies against influenza A viruses bind to the conserved stem region or the receptor-binding cavity of hemagglutinin and the interaction is dominated by the heavy chain. The light chain, however, contributes few or no direct contacts to the antigen. Here we report an H3-clade neutralizing human monoclonal antibody, AF4H1K1, which recognizes the hemagglutinin glycoproteins of all group 2 influenza A viruses. This human monoclonal antibody has been obtained through the screening by pairing different heavy and light chains from an H7N9-infected patient based on the next-generation sequencing technology. Further structural studies revealed that light chains modulate the neutralizing spectrum by affecting the local conformation of heavy chains, instead of direct interaction with the antigen. These findings provide important clues to understand the molecular basis of light chains in antigen recognition and to explore the strategies in particular of the use of light chain modification to develop broadly protective monoclonal antibodies against influenza A viruses and other emerging viruses.
Collapse
Affiliation(s)
- Haixia Xiao
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China
| | - Tianling Guo
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China
| | - Mi Yang
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,3CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| | - Jianxun Qi
- 2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,3CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| | - Chaobin Huang
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China
| | | | - Jinjin Gu
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,5College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457 China
| | - Xuefei Pang
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China
| | - William Jun Liu
- 6National Institute for Viral Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing, 102206 China
| | - Ruchao Peng
- 2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,3CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| | - John McCauley
- 7WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT UK
| | - Yuhai Bi
- 2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,3CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| | - Shihua Li
- 2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,3CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| | - Junxia Feng
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China
| | - Hailiang Zhang
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,5College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457 China
| | - Xupei Zhang
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China
| | - Xishan Lu
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China
| | - Jinghua Yan
- 8CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| | - Liling Chen
- Suzhou Centre for Disease Control and Prevention, Suzhou, 215004 China
| | - Yi Shi
- 2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,3CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| | | | - George Fu Gao
- 1Laboraroty of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, 300308 China.,2CAS Centre for Influenza Research and Early-Warning (CASCIRE), Chinese Academy of Sciences (CAS), China Research Network, Beijing, 100101 China.,3CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101 China
| |
Collapse
|
36
|
Han L, Chen C, Han X, Lin S, Ao X, Han X, Wang J, Ye H. Structural Insights for Anti-Influenza Vaccine Design. Comput Struct Biotechnol J 2019; 17:475-483. [PMID: 31007873 PMCID: PMC6458449 DOI: 10.1016/j.csbj.2019.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/17/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023] Open
Abstract
Influenza A virus are a persistent and significant threat to human health, and current vaccines do not provide sufficient protection due to antigenic drift, which allows influenza viruses to easily escape immune surveillance and antiviral drug activity. Influenza hemagglutinin (HA) is a glycoprotein needed for the entry of enveloped influenza viruses into host cells and is a potential target for anti-influenza humoral immune responses. In recent years, a number of broadly neutralizing antibodies (bnAbs) have been isolated, and their relative structural information obtained from the crystallization of influenza antigens in complex with bnAbs has provided some new insights into future influenza vaccine research. Here, we review the current knowledge of the HA-targeted bnAbs and the structure-based mechanisms contributing to neutralization. We also discuss the potential for this structure-based approach to overcome the challenge of obtaining a highly desired "universal" influenza vaccine, especially on small proteins and peptides.
Collapse
Affiliation(s)
- Lifen Han
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Cong Chen
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Xianlin Han
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Shujin Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
| | - Xiulan Ao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
| | - Xiao Han
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Jianmin Wang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Hanhui Ye
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
| |
Collapse
|
37
|
Abstract
Annually recurring seasonal influenza causes massive economic loss and poses severe threats to public health worldwide. The current seasonal influenza vaccines are the most effective means of preventing influenza infections but possess major weaknesses. Seasonal influenza vaccines require annual updating of the vaccine strains. However, it is an unreachable task to accurately predict the future circulating strains. Vaccines with mismatched strains dramatically compromise the vaccine efficacy. In addition, the seasonal influenza vaccines are ineffective against an unpredictable pandemic. A universal influenza vaccine would overcome these weaknesses of the seasonal vaccines and abolish the threat of influenza pandemics. One approach under investigation is to design influenza vaccine immunogens based on conserved, type-specific amino acid sequences and conformational epitopes, rather than strain-specific. Such vaccines can elicit broadly reactive humoral and cellular immunity. Universal influenza vaccine development has intensively employed nanotechnology because the structural and morphological properties of nanoparticles dramatically improve vaccine immunogenicity and the induced immunity duration. Layered protein nanoparticles can decrease off-target immune responses, fine-tune antigen recognition and processing, and facilitate comprehensive immune response induction. Herein, we review the designs of effective nanoparticle universal influenza vaccines, the recent discoveries of specific nanoparticle features that contribute to immunogenicity enhancement, and recent progress in clinical trials.
Collapse
Affiliation(s)
- Lei Deng
- Center for Inflammation, Immunity & Infection, Georgia State University, 145 Piedmont Avenue SE, Atlanta, Georgia 30302-3965, United States
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, 145 Piedmont Avenue SE, Atlanta, Georgia 30302-3965, United States
| |
Collapse
|
38
|
Sunwoo SY, Schotsaert M, Morozov I, Davis AS, Li Y, Lee J, McDowell C, Meade P, Nachbagauer R, García-Sastre A, Ma W, Krammer F, Richt JA. A Universal Influenza Virus Vaccine Candidate Tested in a Pig Vaccination-Infection Model in the Presence of Maternal Antibodies. Vaccines (Basel) 2018; 6:vaccines6030064. [PMID: 30223475 PMCID: PMC6161263 DOI: 10.3390/vaccines6030064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022] Open
Abstract
The antigenically conserved hemagglutinin stalk region is a target for universal influenza virus vaccines since antibodies against it can provide broad protection against influenza viruses of different subtypes. We tested a universal influenza virus vaccination regimen based on sequential immunization with chimeric hemagglutinin (HA) containing viruses in a swine influenza virus pig model with maternal antibodies against pandemic H1N1. Vaccines were administered as live attenuated virus or inactivated influenza virus split vaccine (+/− Emulsigen adjuvant). As controls, we included groups that received trivalent inactivated influenza vaccine that contained pandemic H1N1 antigens, inactivated adjuvanted H1N2 vaccine (control group for vaccine associated enhanced respiratory disease in the pig model) or mock-vaccination. No induction of H1 head or stalk-specific antibody responses was observed upon vaccination, while responses against H3 and influenza B HA were elicited in the group vaccinated with the trivalent vaccine. Four weeks post vaccination, pigs were intratracheally challenged with pandemic H1N1 virus and euthanized 5 days after challenge. Despite the lack of detectable anti-stalk immunity, the chimeric hemagglutinin vaccine resulted in better clinical outcomes compared to control groups.
Collapse
Affiliation(s)
- Sun-Young Sunwoo
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Igor Morozov
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Anne Sally Davis
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Yuhao Li
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Jinhwa Lee
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Chester McDowell
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Philip Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Wenjun Ma
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Juergen A Richt
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
39
|
Bailey ES, Choi JY, Fieldhouse JK, Borkenhagen LK, Zemke J, Zhang D, Gray GC. The continual threat of influenza virus infections at the human-animal interface: What is new from a one health perspective? EVOLUTION MEDICINE AND PUBLIC HEALTH 2018; 2018:192-198. [PMID: 30210800 PMCID: PMC6128238 DOI: 10.1093/emph/eoy013] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022]
Abstract
This year, in 2018, we mark 100 years since the 1918 influenza pandemic. In the last 100 years, we have expanded our knowledge of public health and increased our ability to detect and prevent influenza; however, we still face challenges resulting from these continually evolving viruses. Today, it is clear that influenza viruses have multiple animal reservoirs (domestic and wild), making infection prevention in humans especially difficult to achieve. With this report, we summarize new knowledge regarding influenza A, B, C and D viruses and their control. We also introduce how a multi-disciplinary One Health approach is necessary to mitigate these threats.
Collapse
Affiliation(s)
- Emily S Bailey
- Duke Global Health Institute, Duke University, Durham, NC, USA.,Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Jessica Y Choi
- Duke Global Health Institute, Duke University, Durham, NC, USA.,Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Jane K Fieldhouse
- Duke Global Health Institute, Duke University, Durham, NC, USA.,Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Laura K Borkenhagen
- Duke Global Health Institute, Duke University, Durham, NC, USA.,Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Juliana Zemke
- Duke Global Health Institute, Duke University, Durham, NC, USA.,Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Dingmei Zhang
- Duke Global Health Institute, Duke University, Durham, NC, USA.,Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Gregory C Gray
- Duke Global Health Institute, Duke University, Durham, NC, USA.,Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.,Global Health Research Center, Duke-Kunshan University, Kunshan, China.,Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore
| |
Collapse
|
40
|
Zoonotic Influenza and Human Health-Part 2: Clinical Features, Diagnosis, Treatment, and Prevention Strategies. Curr Infect Dis Rep 2018; 20:38. [PMID: 30069787 PMCID: PMC7102074 DOI: 10.1007/s11908-018-0643-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Purpose of Review Zoonotic influenza viruses are those influenza viruses that cross the animal-human barrier and can cause disease in humans, manifesting from minor respiratory illnesses to multiorgan dysfunction. The increasing incidence of infections caused by these viruses worldwide has necessitated focused attention to improve both diagnostic as well as treatment modalities. In this second part of a two-part review, we discuss the clinical features, diagnostic modalities, and treatment of zoonotic influenza, and provide an overview of prevention strategies. Recent Findings Illnesses caused by novel reassortant avian influenza viruses continue to be detected and described; most recently, a human case of avian influenza A(H7N4) has been described from China. We continue to witness increasing rates of A(H7N9) infections, with the latest (fifth) wave, from late 2016 to 2017, being the largest to date. The case fatality rate for A(H7N9) and A(H5N1) infections among humans is much higher than that of seasonal influenza infections. Since the emergence of the A(H1N1) 2009 pandemic, and subsequently A(H7N9), testing and surveillance for novel influenzas have become more effective. Various newer treatment options, including peramivir, favipiravir (T-705), and DAS181, and human or murine monoclonal antibodies have been evaluated in vitro and in animal models. Summary Armed with robust diagnostic modalities, antiviral medications, vaccines, and advanced surveillance systems, we are today better prepared to face a new influenza pandemic and to limit the burden of zoonotic influenza than ever before. Sustained efforts and robust research are necessary to efficiently deal with the highly mutagenic zoonotic influenza viruses.
Collapse
|
41
|
Nogales A, Piepenbrink MS, Wang J, Ortega S, Basu M, Fucile CF, Treanor JJ, Rosenberg AF, Zand MS, Keefer MC, Martinez-Sobrido L, Kobie JJ. A Highly Potent and Broadly Neutralizing H1 Influenza-Specific Human Monoclonal Antibody. Sci Rep 2018. [PMID: 29531320 PMCID: PMC5847613 DOI: 10.1038/s41598-018-22307-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Influenza’s propensity for antigenic drift and shift, and to elicit predominantly strain specific antibodies (Abs) leaves humanity susceptible to waves of new strains with pandemic potential for which limited or no immunity may exist. Subsequently new clinical interventions are needed. To identify hemagglutinin (HA) epitopes that if targeted may confer universally protective humoral immunity, we examined plasmablasts from a subject that was immunized with the seasonal influenza inactivated vaccine, and isolated a human monoclonal Ab (mAb), KPF1. KPF1 has broad and potent neutralizing activity against H1 influenza viruses, and recognized 83% of all H1 isolates tested, including the pandemic 1918 H1. Prophylactically, KPF1 treatment resulted in 100% survival of mice from lethal challenge with multiple H1 influenza strains and when given as late as 72 h after challenge with A/California/04/2009 H1N1, resulted in 80% survival. KPF1 recognizes a novel epitope in the HA globular head, which includes a highly conserved amino acid, between the Ca and Cb antigenic sites. Although recent HA stalk-specific mAbs have broader reactivity, their potency is substantially limited, suggesting that cocktails of broadly reactive and highly potent HA globular head-specific mAbs, like KPF1, may have greater clinical feasibility for the treatment of influenza infections.
Collapse
Affiliation(s)
- Aitor Nogales
- Department of Microbiology & Immunology, University of Rochester, Rochester, NY, USA
| | | | - Jiong Wang
- Division of Nephrology, University of Rochester, Rochester, NY, USA
| | - Sandra Ortega
- Department of Microbiology & Immunology, University of Rochester, Rochester, NY, USA
| | - Madhubanti Basu
- Infectious Diseases Division, University of Rochester, Rochester, NY, USA
| | - Christopher F Fucile
- Department of Microbiology, Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John J Treanor
- Infectious Diseases Division, University of Rochester, Rochester, NY, USA
| | - Alexander F Rosenberg
- Department of Microbiology, Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin S Zand
- Division of Nephrology, University of Rochester, Rochester, NY, USA
| | - Michael C Keefer
- Infectious Diseases Division, University of Rochester, Rochester, NY, USA
| | - Luis Martinez-Sobrido
- Department of Microbiology & Immunology, University of Rochester, Rochester, NY, USA.
| | - James J Kobie
- Infectious Diseases Division, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
42
|
Double-layered protein nanoparticles induce broad protection against divergent influenza A viruses. Nat Commun 2018; 9:359. [PMID: 29367723 PMCID: PMC5783933 DOI: 10.1038/s41467-017-02725-4] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/20/2017] [Indexed: 01/22/2023] Open
Abstract
Current influenza vaccines provide limited protection against circulating influenza A viruses. A universal influenza vaccine will eliminate the intrinsic limitations of the seasonal flu vaccines. Here we report methodology to generate double-layered protein nanoparticles as a universal influenza vaccine. Layered nanoparticles are fabricated by desolvating tetrameric M2e into protein nanoparticle cores and coating these cores by crosslinking headless HAs. Representative headless HAs of two HA phylogenetic groups are constructed and purified. Vaccinations with the resulting protein nanoparticles in mice induces robust long-lasting immunity, fully protecting the mice against challenges by divergent influenza A viruses of the same group or both groups. The results demonstrate the importance of incorporating both structure-stabilized HA stalk domains and M2e into a universal influenza vaccine to improve its protective potency and breadth. These potent disassemblable protein nanoparticles indicate a wide application in protein drug delivery and controlled release. Relatively well conserved domains of influenza A virus (IAV) proteins are potential candidates for the development of a universal IAV vaccine. Here, Deng et al. combine two such conserved antigens (M2e and HA stalk) in a double-layered protein nanoparticle and show that it protects against divergent IAVs in mice.
Collapse
|
43
|
McKimm-Breschkin JL, Jiang S, Hui DS, Beigel JH, Govorkova EA, Lee N. Prevention and treatment of respiratory viral infections: Presentations on antivirals, traditional therapies and host-directed interventions at the 5th ISIRV Antiviral Group conference. Antiviral Res 2018; 149:118-142. [PMID: 29162476 PMCID: PMC7133686 DOI: 10.1016/j.antiviral.2017.11.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
Abstract
The International Society for Influenza and other Respiratory Virus Diseases held its 5th Antiviral Group (isirv-AVG) Conference in Shanghai, China, in conjunction with the Shanghai Public Health Center and Fudan University from 14-16 June 2017. The three-day programme encompassed presentations on some of the clinical features, management, immune responses and virology of respiratory infections, including influenza A(H1N1)pdm09 and A(H7N9) viruses, MERS-CoV, SARS-CoV, adenovirus Type 80, enterovirus D68, metapneumovirus and respiratory syncytial virus (RSV). Updates were presented on several therapeutics currently in clinical trials, including influenza polymerase inhibitors pimodivir/JNJ6362387, S033188, favipiravir, monoclonal antibodies MHAA45449A and VIS410, and host directed strategies for influenza including nitazoxanide, and polymerase ALS-008112 and fusion inhibitors AK0529, GS-5806 for RSV. Updates were also given on the use of the currently licensed neuraminidase inhibitors. Given the location in China, there were also presentations on the use of Traditional Chinese Medicines. Following on from the previous conference, there were ongoing discussions on appropriate endpoints for severe influenza in clinical trials from regulators and clinicians, an issue which remains unresolved. The aim of this conference summary is to provide information for not only conference participants, but a detailed referenced review of the current status of clinical trials, and pre-clinical development of therapeutics and vaccines for influenza and other respiratory diseases for a broader audience.
Collapse
Affiliation(s)
| | - Shibo Jiang
- College of Basic Medical Sciences, Fudan University, Shanghai, China; Lindsley F. Kimball Research Institute, New York Blood Center, NY, USA
| | - David S Hui
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - John H Beigel
- Leidos Biomedical Research, Inc., Support to National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, USA
| | - Nelson Lee
- Faculty of Medicine and Dentistry, University of Alberta, Canada
| |
Collapse
|
44
|
A Potent Germline-like Human Monoclonal Antibody Targets a pH-Sensitive Epitope on H7N9 Influenza Hemagglutinin. Cell Host Microbe 2017; 22:471-483.e5. [PMID: 28966056 PMCID: PMC6290738 DOI: 10.1016/j.chom.2017.08.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/12/2017] [Accepted: 08/22/2017] [Indexed: 11/20/2022]
Abstract
The H7N9 influenza virus causes high-mortality disease in humans but no effective therapeutics are available. Here we report a human monoclonal antibody, m826, that binds to H7 hemagglutinin (HA) and protects against H7N9 infection. m826 binds to H7N9 HA with subnanomolar affinity at acidic pH and 10-fold lower affinity at neutral pH. The high-resolution (1.9 Å) crystal structure of m826 complexed with H7N9 HA indicates that m826 binds an epitope that may be fully exposed upon pH-induced conformational changes in HA. m826 fully protects mice against lethal challenge with H7N9 virus through mechanisms likely involving antibody-dependent cell-mediated cytotoxicity. Interestingly, immunogenetic analysis indicates that m826 is a germline antibody, and m826-like sequences can be identified in H7N9-infected patients, healthy adults, and newborn babies. These m826 properties offer a template for H7N9 vaccine immunogens, a promising candidate therapeutic, and a tool for exploring mechanisms of virus infection inhibition by antibodies.
Collapse
|
45
|
Pierce CL, Williams TL, Santana WI, Levine M, Chen LM, Cooper HC, Solano MI, Woolfitt AR, Marasco WA, Fang H, Donis RO, Barr JR. Immunocapture isotope dilution mass spectrometry in response to a pandemic influenza threat. Vaccine 2017; 35:5011-5018. [PMID: 28774565 PMCID: PMC11235095 DOI: 10.1016/j.vaccine.2017.07.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/12/2017] [Accepted: 07/16/2017] [Indexed: 01/02/2023]
Abstract
As a result of recent advances in mass spectrometry-based protein quantitation methods, these techniques are now poised to play a critical role in rapid formulation of pandemic influenza vaccines. Analytical techniques that have been developed and validated on seasonal influenza strains can be used to increase the quality and decrease the time required to deliver protective pandemic vaccines to the global population. The emergence of a potentially pandemic avian influenza A (H7N9) virus in March of 2013, prompted the US public health authorities and the vaccine industry to initiate production of a pre-pandemic vaccine for preparedness purposes. To this end, we evaluated the feasibility of using immunocapture isotope dilution mass spectrometry (IC-IDMS) to evaluate the suitability of the underlying monoclonal and polyclonal antibodies (mAbs and pAbs) for their capacity to isolate the H7 hemagglutinin (HA) in this new vaccine for quantification by IDMS. A broad range of H7 capture efficiencies was observed among mAbs tested by IC-IDMS with FR-545, 46/6, and G3 A533 exhibiting the highest cross-reactivity capabilities to H7 of A/Shanghai/2/2013. MAb FR-545 was selected for continued assessment, evaluated by IC-IDMS for mAb reactivity against H7 in the H7N9 candidate vaccine virus and compared with/to reactivity to the reference polyclonal antiserum in allantoic fluid, purified whole virus, lyophilized whole virus and final detergent-split monovalent vaccine preparations for vaccine development. IC-IDMS assessment of FR-545 alongside IC-IDMS using the reference polyclonal antiserum to A/Shanghai/2/2013 and with the regulatory SRID method showed strong correlation and mAb IC-IDMS could have played an important role in the event a potential surrogate potency test was required to be rapidly implemented.
Collapse
Affiliation(s)
- Carrie L Pierce
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Tracie L Williams
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Wanda I Santana
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Marnie Levine
- Battelle Memorial Institute, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Li-Mei Chen
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - Hans C Cooper
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Maria I Solano
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Adrian R Woolfitt
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Wayne A Marasco
- Department of Cancer Immunology & Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - He Fang
- Institute of Preventive Veterinary Medicine and Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China.
| | - Ruben O Donis
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA.
| | - John R Barr
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| |
Collapse
|
46
|
Tyrrell BE, Sayce AC, Warfield KL, Miller JL, Zitzmann N. Iminosugars: Promising therapeutics for influenza infection. Crit Rev Microbiol 2017; 43:521-545. [PMID: 27931136 PMCID: PMC5470110 DOI: 10.1080/1040841x.2016.1242868] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/02/2016] [Accepted: 09/27/2016] [Indexed: 01/11/2023]
Abstract
Influenza virus causes three to five million severe respiratory infections per year in seasonal epidemics, and sporadic pandemics, three of which occurred in the twentieth century and are a continuing global threat. Currently licensed antivirals exclusively target the viral neuraminidase or M2 ion channel, and emerging drug resistance necessitates the development of novel therapeutics. It is believed that a host-targeted strategy may combat the development of antiviral drug resistance. To this end, a class of molecules known as iminosugars, hydroxylated carbohydrate mimics with the endocyclic oxygen atom replaced by a nitrogen atom, are being investigated for their broad-spectrum antiviral potential. The influenza virus glycoproteins, hemagglutinin and neuraminidase, are susceptible to inhibition of endoplasmic reticulum α-glucosidases by certain iminosugars, leading to reduced virion production or infectivity, demonstrated by in vitro and in vivo studies. In some experiments, viral strain-specific effects are observed. Iminosugars may also inhibit other host and virus targets with antiviral consequences. While investigations of anti-influenza iminosugar activities have been conducted since the 1980s, recent successes of nojirimycin derivatives have re-invigorated investigation of the therapeutic potential of iminosugars as orally available, low cytotoxicity, effective anti-influenza drugs.
Collapse
Affiliation(s)
- Beatrice Ellen Tyrrell
- Department of Biochemistry, University of Oxford Medical Sciences DivisionOxfordUnited Kingdom of Great Britain and Northern Ireland
| | - Andrew Cameron Sayce
- Department of Biochemistry, University of Oxford Medical Sciences DivisionOxfordUnited Kingdom of Great Britain and Northern Ireland
| | - Kelly Lyn Warfield
- Antiviral Research and Development, Emergent BioSolutions IncGaithersburgMDUnited States
| | - Joanna Louise Miller
- Department of Biochemistry, University of Oxford Medical Sciences DivisionOxfordUnited Kingdom of Great Britain and Northern Ireland
| | - Nicole Zitzmann
- Department of Biochemistry, University of Oxford Medical Sciences DivisionOxfordUnited Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
47
|
Konevtsova OV, Pimonov VV, Lorman VL, Rochal SB. Quasicrystalline and crystalline types of local protein order in capsids of small viruses. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2017; 29:284002. [PMID: 28488589 DOI: 10.1088/1361-648x/aa7211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Like metal alloys and micellar systems in soft matter, the viral capsid structures can be of crystalline and quasicrystalline types. We reveal the local quasicrystalline order of proteins in small spherical viral capsids using their nets of dodecahedral type. We show that the structure of some of the viral shells is well described in terms of a chiral pentagonal tiling, whose nodes coincide with centers of mass of protein molecules. The chiral protein packing found in these capsids originates from the pentagonal Penrose tiling (PPT), due to a specific phason reconstruction needed to fit the protein order at the adjacent dodecahedron faces. Via examples of small spherical viral shells and geminate capsid of a Maize Streak virus, we discuss the benefits and shortcomings of the usage of a dodecahedral net in comparison to icosahedral one, which is commonly applied for the modeling of viral shells with a crystalline local order.
Collapse
Affiliation(s)
- O V Konevtsova
- Faculty of Physics, Southern Federal University, 5 Zorge Str., 344090 Rostov-on-Don, Russia
| | | | | | | |
Collapse
|
48
|
Su S, Gu M, Liu D, Cui J, Gao GF, Zhou J, Liu X. Epidemiology, Evolution, and Pathogenesis of H7N9 Influenza Viruses in Five Epidemic Waves since 2013 in China. Trends Microbiol 2017; 25:713-728. [PMID: 28734617 DOI: 10.1016/j.tim.2017.06.008] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 01/30/2023]
Abstract
H7N9 influenza viruses were first isolated in 2013 and continue to cause human infections. H7N9 infections represent an ongoing public health threat that has resulted in 1344 cases with 511 deaths as of April 9, 2017. This highlights the continued threat posed by the current poultry trade and live poultry market system in China. Until now, there have been five H7N9 influenza epidemic waves in China; however, the steep increase in the number of humans infected with H7N9 viruses observed in the fifth wave, beginning in October 2016, the spread into western provinces, and the emergence of highly pathogenic (HP) H7N9 influenza outbreaks in chickens and infection in humans have caused domestic and international concern. In this review, we summarize and compare the different waves of H7N9 regarding their epidemiology, pathogenesis, evolution, and characteristic features, and speculate on factors behind the recent increase in the number of human cases and sudden outbreaks in chickens. The continuous evolution of the virus poses a long-term threat to public health and the poultry industry, and thus it is imperative to strengthen prevention and control strategies.
Collapse
Affiliation(s)
- Shuo Su
- Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Min Gu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Di Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jie Cui
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, China
| | - Jiyong Zhou
- Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University, Hangzhou 310058, China; Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China.
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, 225009, China; Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
49
|
Danqi B, Li Z, Liu Q, Richt JA. H7N9 avian influenza A virus in China: a short report on its circulation, drug resistant mutants and novel antiviral drugs. Expert Rev Anti Infect Ther 2017; 15:723-727. [PMID: 28692316 DOI: 10.1080/14787210.2017.1353419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The first human H7N9 avian influenza virus case was reported in Shanghai in 2013. Shortly thereafter, this virus spread to other regions in China. Molecular analysis indicated that the H7N9 virus is a reassortant virus containing internal genes from the H9N2 virus and previously described mammalian adaption markers, which could allow the virus to adapt efficiently to a mammalian host. Fortunately, there is no evidence of sustained person-to-person spread. Most of the human H7N9 cases have a history of exposure to live poultry markets (LPMs). The circulating H7N9 were low pathogenic viruses, however highly pathogenic H7N9 viruses were recently identified in human cases. Areas covered: In the present article, the circulation of H7N9 in LPMs of China, the five waves of H7N9 infection in humans, recently identified drug resistant mutants and potential antiviral drugs against H7N9 are discussed; this may provide further understanding of the evolution and pandemic potential of the H7N9 influenza viruses. Expert commentary: All the data reveal that the major source of H7N9 viruses are LPMs and the H7N9 virus is still circulating widely in China. It is concerning that the recent emergence of highly pathogenic H7N9 viruses may result in highly transmissible viruses in mammalian species.
Collapse
Affiliation(s)
- Bao Danqi
- a Department of Avian Diseases , Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Shanghai , People's Republic of China.,b College of Veterinary Medicine, Inner Mongolia Agricultural University , Hohhot , People's Republic of China
| | - Zejun Li
- a Department of Avian Diseases , Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Shanghai , People's Republic of China
| | - Qinfang Liu
- a Department of Avian Diseases , Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Shanghai , People's Republic of China
| | - Juergen A Richt
- c Diagnostic Medicine/Pathobiology , College of Veterinary Medicine, Kansas State University , Manhattan , KS , USA
| |
Collapse
|
50
|
Corti D, Cameroni E, Guarino B, Kallewaard NL, Zhu Q, Lanzavecchia A. Tackling influenza with broadly neutralizing antibodies. Curr Opin Virol 2017; 24:60-69. [PMID: 28527859 PMCID: PMC7102826 DOI: 10.1016/j.coviro.2017.03.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/02/2017] [Accepted: 03/09/2017] [Indexed: 11/28/2022]
Abstract
Monoclonal antibodies have revolutionized the treatment of several human diseases, including cancer, autoimmunity and inflammatory conditions and represent a new frontier for the treatment of infectious diseases. In the last decade, new methods have allowed the efficient interrogation of the human antibody repertoire from influenza immune individuals and the isolation of several monoclonal antibodies capable of dealing with the high variability of influenza viruses. Here, we will provide a comprehensive overview of the specificity, antiviral and immunological mechanisms of action and development into the clinic of broadly reactive monoclonal antibodies against influenza A and B viruses.
Collapse
Affiliation(s)
| | | | | | | | - Qing Zhu
- Department of Infectious Disease, MedImmune, Gaithersburg, MD 20878, USA
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| |
Collapse
|