1
|
Johnson VJ, Luster MI, Edwards A, Kashon M, Burleson GR, Burleson FG, Germolec DR. An In Vitro test battery using human whole blood for immunotoxicity hazard identification: Proof of concept studies with dexamethasone and benzo(a)pyrene. Toxicology 2025; 515:154153. [PMID: 40233920 DOI: 10.1016/j.tox.2025.154153] [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: 03/07/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025]
Abstract
Immunotoxicity assessment is nearing a crossroads predicated on mounting pressure for reduction/replacement of animals in toxicology. This has fueled the development of alternative New Approach Methodologies (NAMs) for hazard identification. This work details a comprehensive human whole blood NAM battery for immunotoxicity testing. The test system is a closed tube culture containing whole blood diluted 1:3 in culture media with or without an immune stimulant, anti-CD3/CD28 or viral peptide pool. Model immunotoxicants dexamethasone (DEX; 0 - 1.94 μM) and benzo(a)pyrene [B(a)P; 0-6.3 μM], were added to the test system for 24 hours. Immune cells were identified and counted by flow cytometric immunophenotyping and assessed for natural killer (NK) cell activity and T cell activation. Supernatants were interrogated for proinflammatory cytokine concentrations. In vitro treatment with DEX resulted in concentration-dependent suppression of cytokine production, NK cell activity, and T cell activation induced by anti-CD3/CD28, as well as viral-induced cytokine production. B(a)P caused suppression of cytokine production and a nonsignificant reduction in T cell activation but did not impact NK cell activity, however, immunosuppression by B(a)P only occurred following metabolic activation by S9 fraction enzymes. Advantages of this NAM battery include assessment of adaptive immunity and direct translation of immunotoxicity to viral host resistance. These results provide evidence of in vitro immunotoxicity that reflect known outcomes from in vivo studies. This multi-endpoint human whole blood NAM battery should be useful for screening compounds for immunotoxicity hazard identification without reliance on animal systems for increased translatability to humans.
Collapse
Affiliation(s)
| | | | - Amber Edwards
- Burleson Research Technologies, Inc, Morrisville, NC, USA
| | - Michael Kashon
- Burleson Research Technologies, Inc, Morrisville, NC, USA; Mountainview Research Analytics, Morgantown, WV, USA
| | | | | | - Dori R Germolec
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| |
Collapse
|
2
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Jyothi SR, Kundlas M, Joshi KK, Gulyamov S, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. From protein to immunology: comprehensive insights into Marburg virus vaccines, mechanism, and application. Arch Microbiol 2025; 207:74. [PMID: 40025302 DOI: 10.1007/s00203-025-04277-4] [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: 01/02/2025] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
The Marburg virus (MARV), a member of the Filoviridae family, is a highly lethal pathogen that causes Marburg virus disease (MVD), a severe hemorrhagic fever with high fatality rates.Despite recurrent outbreaks, no licensed vaccine is currently available. This review explores MARV's genomic architecture, structural proteins, and recent advancements in vaccine development. It highlights the crucial role of MARV's seven monocistronic genes in viral replication and pathogenesis, with a focus on structural proteins such as nucleoprotein (NP), glycoprotein (GP), and viral proteins VP35, VP40, and VP24. These proteins are essential for viral entry, immune evasion, and replication. The review further examines various vaccine platforms, including multi-epitope vaccines, DNA-based vaccines, viral vector vaccines, virus-like particles (VLPs), and mRNA vaccines. Cutting-edge immunoinformatics approaches are discussed for identifying conserved epitopes critical for broad-spectrum protection. The immunological responses induced by these vaccine candidates, particularly their efficacy in preclinical trials, are analyzed, showcasing promising results in generating both humoral and cellular immunity. Moreover, the review addresses challenges and future directions in MARV vaccine development, emphasizing the need for enhanced immunogenicity, safety, and global accessibility. The integration of omics technologies (genomics, transcriptomics, proteomics) with immunoinformatics is presented as a transformative approach for next-generation vaccine design. Innovative platforms such as mRNA and VLP-based vaccines offer rapid and effective development opportunities. In this study, underscores the urgent need for a licensed MARV vaccine to prevent future outbreaks and strengthen global preparedness. By synthesizing the latest research and technological advancements, it provides a strategic roadmap for developing safe, effective, and broadly protective vaccines. The fight against MARV is a global priority, requiring coordinated efforts from researchers, policymakers, and public health organizations.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, Uttarakhand, 248002, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Surat Gulyamov
- Department of Dentistry and Pediatric Dentistry, Tashkent Pediatric Medical Institute, Bogishamol Street 223, 100140, Tashkent, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
3
|
Amoah S, Cao W, Sayedahmed EE, Wang Y, Kumar A, Mishina M, Eddins DJ, Wang WC, Burroughs M, Sheth M, Lee J, Shieh WJ, Ray SD, Bohannon CD, Ranjan P, Sharma SD, Hoehner J, Arthur RA, Gangappa S, Wakamatsu N, Johnston HR, Pohl J, Mittal SK, Sambhara S. The frequency and function of nucleoprotein-specific CD8 + T cells are critical for heterosubtypic immunity against influenza virus infection. J Virol 2024; 98:e0071124. [PMID: 39082839 PMCID: PMC11334528 DOI: 10.1128/jvi.00711-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) mediate host defense against viral and intracellular bacterial infections and tumors. However, the magnitude of CTL response and their function needed to confer heterosubtypic immunity against influenza virus infection are unknown. We addressed the role of CD8+ T cells in the absence of any cross-reactive antibody responses to influenza viral proteins using an adenoviral vector expressing a 9mer amino acid sequence recognized by CD8+ T cells. Our results indicate that both CD8+ T cell frequency and function are crucial for heterosubtypic immunity. Low morbidity, lower viral lung titers, low to minimal lung pathology, and better survival upon heterosubtypic virus challenge correlated with the increased frequency of NP-specific CTLs. NP-CD8+ T cells induced by differential infection doses displayed distinct RNA transcriptome profiles and functional properties. CD8+ T cells induced by a high dose of influenza virus secreted significantly higher levels of IFN-γ and exhibited higher levels of cytotoxic function. The mice that received NP-CD8+ T cells from the high-dose virus recipients through adoptive transfer had lower viral titers following viral challenge than those induced by the low dose of virus, suggesting differential cellular programming by antigen dose. Enhanced NP-CD8+ T-cell functions induced by a higher dose of influenza virus strongly correlated with the increased expression of cellular and metabolic genes, indicating a shift to a more glycolytic metabolic phenotype. These findings have implications for developing effective T cell vaccines against infectious diseases and cancer. IMPORTANCE Cytotoxic T lymphocytes (CTLs) are an important component of the adaptive immune system that clears virus-infected cells or tumor cells. Hence, developing next-generation vaccines that induce or recall CTL responses against cancer and infectious diseases is crucial. However, it is not clear if the frequency, function, or both are essential in conferring protection, as in the case of influenza. In this study, we demonstrate that both CTL frequency and function are crucial for providing heterosubtypic immunity to influenza by utilizing an Ad-viral vector expressing a CD8 epitope only to rule out the role of antibodies, single-cell RNA-seq analysis, as well as adoptive transfer experiments. Our findings have implications for developing T cell vaccines against infectious diseases and cancer.
Collapse
Affiliation(s)
- Samuel Amoah
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Weiping Cao
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, 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
| | - Amrita Kumar
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Margarita Mishina
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Devon J. Eddins
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wen-Chien Wang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Mark Burroughs
- 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
| | - Mili Sheth
- 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
| | - Justin Lee
- 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
| | - Wun-Ju Shieh
- Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sean D. Ray
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Caitlin D. Bohannon
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Priya Ranjan
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Suresh D. Sharma
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jessica Hoehner
- Emory Integrated Computational Core, Emory Integrated Core Facilities, Emory University, Atlanta, Georgia, USA
| | - Robert A. Arthur
- Emory Integrated Computational Core, Emory Integrated Core Facilities, Emory University, Atlanta, Georgia, USA
| | - Shivaprakash Gangappa
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nobuko Wakamatsu
- Indiana Animal Disease Diagnostic Laboratory, Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - H. Richard Johnston
- Department of Human Genetics, Emory University School of Medicine, Emory University, Atlanta, Georgia, 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
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Tran KA, Pernet E, Sadeghi M, Downey J, Chronopoulos J, Lapshina E, Tsai O, Kaufmann E, Ding J, Divangahi M. BCG immunization induces CX3CR1 hi effector memory T cells to provide cross-protection via IFN-γ-mediated trained immunity. Nat Immunol 2024; 25:418-431. [PMID: 38225437 DOI: 10.1038/s41590-023-01739-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
After a century of using the Bacillus Calmette-Guérin (BCG) vaccine, our understanding of its ability to provide protection against homologous (Mycobacterium tuberculosis) or heterologous (for example, influenza virus) infections remains limited. Here we show that systemic (intravenous) BCG vaccination provides significant protection against subsequent influenza A virus infection in mice. We further demonstrate that the BCG-mediated cross-protection against influenza A virus is largely due to the enrichment of conventional CD4+ effector CX3CR1hi memory αβ T cells in the circulation and lung parenchyma. Importantly, pulmonary CX3CR1hi T cells limit early viral infection in an antigen-independent manner via potent interferon-γ production, which subsequently enhances long-term antimicrobial activity of alveolar macrophages. These results offer insight into the unknown mechanism by which BCG has persistently displayed broad protection against non-tuberculosis infections via cross-talk between adaptive and innate memory responses.
Collapse
Affiliation(s)
- Kim A Tran
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Erwan Pernet
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Medical Biology, Université du Québec à Trois-Rivières, Quebec, Quebec, Canada
| | - Mina Sadeghi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Jeffrey Downey
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Julia Chronopoulos
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Elizabeth Lapshina
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Oscar Tsai
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Eva Kaufmann
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Ding
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Maziar Divangahi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
5
|
Ortiz JR, Bernstein DI, Hoft DF, Woods CW, McClain MT, Frey SE, Brady RC, Bryant C, Wegel A, Frenck RW, Walter EB, Abate G, Williams SR, Atmar RL, Keitel WA, Rouphael N, Memoli MJ, Makhene MK, Roberts PC, Neuzil KM. A Multicenter, Controlled Human Infection Study of Influenza A(H1N1)pdm09 in Healthy Adults. J Infect Dis 2023; 228:287-298. [PMID: 36702771 PMCID: PMC10420403 DOI: 10.1093/infdis/jiad021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND We evaluated the associations between baseline influenza virus-specific hemagglutination inhibition (HAI) and microneutralization (MN) titers and subsequent symptomatic influenza virus infection in a controlled human infection study. METHODS We inoculated unvaccinated healthy adults aged 18-49 years with an influenza A/California/04/2009/H1N1pdm-like virus (NCT04044352). We collected serial safety labs, serum for HAI and MN, and nasopharyngeal swabs for reverse-transcription polymerase chain reaction (RT-PCR) testing. Analyses used the putative seroprotective titer of ≥40 for HAI and MN. The primary clinical outcome was mild-to-moderate influenza disease (MMID), defined as ≥1 postchallenge positive qualitative RT-PCR test with a qualifying symptom/clinical finding. RESULTS Of 76 participants given influenza virus challenge, 54 (71.1%) experienced MMID. Clinical illness was generally very mild. MMID attack rates among participants with baseline titers ≥40 by HAI and MN were 64.9% and 67.9%, respectively, while MMID attack rates among participants with baseline titers <40 by HAI and MN were 76.9% and 78.3%, respectively. The estimated odds of developing MMID decreased by 19% (odds ratio, 0.81 [95% confidence interval, .62-1.06]; P = .126) for every 2-fold increase in baseline HAI. There were no significant adverse events. CONCLUSIONS We achieved a 71.1% attack rate of MMID. High baseline HAI and MN were associated with protection from illness. Clinical Trials Registration. NCT04044352.
Collapse
Affiliation(s)
- Justin R Ortiz
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| | - David I Bernstein
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Daniel F Hoft
- Internal Medicine and
- Molecular Microbiology and Immunology, Division of Infectious Diseases, Allergy and Immunology and Center for Vaccine Development, Saint Louis University School of Medicine, Missouri
| | - Christopher W Woods
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | - Micah T McClain
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina
| | | | - Rebecca C Brady
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Christopher Bryant
- Vaccine and Infectious Disease Therapeutic Research Unit, The Emmes Company, Rockville, Maryland
| | - Ashley Wegel
- Vaccine and Infectious Disease Therapeutic Research Unit, The Emmes Company, Rockville, Maryland
| | - Robert W Frenck
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Ohio; Departments of
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina
| | | | - Sarah R Williams
- Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore
| | - Robert L Atmar
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Wendy A Keitel
- Departments of Molecular Virology & Microbiology and Medicine, Baylor College of Medicine, Houston, Texas
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | | | - Mamodikoe K Makhene
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
6
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringuist R, Smith C, Ochoa MA, Roy K. A multiadjuvant polysaccharide-amino acid-lipid (PAL) subunit nanovaccine generates robust systemic and lung-specific mucosal immune responses against SARS-CoV-2 in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539395. [PMID: 37215018 PMCID: PMC10197586 DOI: 10.1101/2023.05.05.539395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, which are essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters could overcome the shortcomings of parenteral vaccines and enhance pre- existing systemic immunity. Here we present a new protein subunit nanovaccine using multiadjuvanted (e.g. RIG-I: PUUC, TLR9: CpG) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL- NPs, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lung, and showed robust systemic humoral immunity. Interestingly, as a purely intranasal vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. Our data suggest that PUUC+CpG PAL-NP subunit vaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
|
7
|
Martins de Camargo M, Caetano AR, Ferreira de Miranda Santos IK. Evolutionary pressures rendered by animal husbandry practices for avian influenza viruses to adapt to humans. iScience 2022; 25:104005. [PMID: 35313691 PMCID: PMC8933668 DOI: 10.1016/j.isci.2022.104005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Commercial poultry operations produce and crowd billions of birds every year, which is a source of inexpensive animal protein. Commercial poultry is intensely bred for desirable production traits, and currently presents very low variability at the major histocompatibility complex. This situation dampens the advantages conferred by the MHC’s high genetic variability, and crowding generates immunosuppressive stress. We address the proteins of influenza A viruses directly and indirectly involved in host specificities. We discuss how mutants with increased virulence and/or altered host specificity may arise if few class I alleles are the sole selective pressure on avian viruses circulating in immunocompromised poultry. This hypothesis is testable with peptidomics of MHC ligands. Breeding strategies for commercial poultry can easily and inexpensively include high variability of MHC as a trait of interest, to help save billions of dollars as a disease burden caused by influenza and decrease the risk of selecting highly virulent strains.
Collapse
|
8
|
Šantak M, Matić Z. The Role of Nucleoprotein in Immunity to Human Negative-Stranded RNA Viruses—Not Just Another Brick in the Viral Nucleocapsid. Viruses 2022; 14:v14030521. [PMID: 35336928 PMCID: PMC8955406 DOI: 10.3390/v14030521] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Negative-stranded RNA viruses (NSVs) are important human pathogens, including emerging and reemerging viruses that cause respiratory, hemorrhagic and other severe illnesses. Vaccine design traditionally relies on the viral surface glycoproteins. However, surface glycoproteins rarely elicit effective long-term immunity due to high variability. Therefore, an alternative approach is to include conserved structural proteins such as nucleoprotein (NP). NP is engaged in myriad processes in the viral life cycle: coating and protection of viral RNA, regulation of transcription/replication processes and induction of immunosuppression of the host. A broad heterosubtypic T-cellular protection was ascribed very early to this protein. In contrast, the understanding of the humoral immunity to NP is very limited in spite of the high titer of non-neutralizing NP-specific antibodies raised upon natural infection or immunization. In this review, the data with important implications for the understanding of the role of NP in the immune response to human NSVs are revisited. Major implications of the elicited T-cell immune responses to NP are evaluated, and the possible multiple mechanisms of the neglected humoral response to NP are discussed. The intention of this review is to remind that NP is a very promising target for the development of future vaccines.
Collapse
|
9
|
Singleton EV, Gates CJ, David SC, Hirst TR, Davies JB, Alsharifi M. Enhanced Immunogenicity of a Whole-Inactivated Influenza A Virus Vaccine Using Optimised Irradiation Conditions. Front Immunol 2021; 12:761632. [PMID: 34899711 PMCID: PMC8652198 DOI: 10.3389/fimmu.2021.761632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus presents a constant pandemic threat due to the mutagenic nature of the virus and the inadequacy of current vaccines to protect against emerging strains. We have developed a whole-inactivated influenza vaccine using γ-irradiation (γ-Flu) that can protect against both vaccine-included strains as well as emerging pandemic strains. γ-irradiation is a widely used inactivation method and several γ-irradiated vaccines are currently in clinical or pre-clinical testing. To enhance vaccine efficacy, irradiation conditions should be carefully considered, particularly irradiation temperature. Specifically, while more damage to virus structure is expected when using higher irradiation temperatures, reduced radiation doses will be required to achieve sterility. In this study, we compared immunogenicity of γ-Flu irradiated at room temperature, chilled on ice or frozen on dry ice using different doses of γ-irradiation to meet internationally accepted sterility assurance levels. We found that, when irradiating at sterilising doses, the structural integrity and vaccine efficacy were well maintained in all preparations regardless of irradiation temperature. In fact, using a higher temperature and lower radiation dose appeared to induce higher neutralising antibody responses and more effective cytotoxic T cell responses. This outcome is expected to simplify irradiation protocols for manufacturing of highly effective irradiated vaccines.
Collapse
Affiliation(s)
- Eve Victoria Singleton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Chloe Jayne Gates
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Shannon Christa David
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Timothy Raymond Hirst
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
- Gamma Vaccines Pty Ltd, Yarralumla, ACT, Australia
| | - Justin Bryan Davies
- Irradiations Group, Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia
| | - Mohammed Alsharifi
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Sciences, University of Adelaide, Adelaide, SA, Australia
- Gamma Vaccines Pty Ltd, Yarralumla, ACT, Australia
| |
Collapse
|
10
|
Calzas C, Mao M, Turpaud M, Viboud Q, Mettier J, Figueroa T, Bessière P, Mangin A, Sedano L, Hervé PL, Volmer R, Ducatez MF, Bourgault S, Archambault D, Le Goffic R, Chevalier C. Immunogenicity and Protective Potential of Mucosal Vaccine Formulations Based on Conserved Epitopes of Influenza A Viruses Fused to an Innovative Ring Nanoplatform in Mice and Chickens. Front Immunol 2021; 12:772550. [PMID: 34868036 PMCID: PMC8632632 DOI: 10.3389/fimmu.2021.772550] [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: 09/08/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Current inactivated vaccines against influenza A viruses (IAV) mainly induce immune responses against highly variable epitopes across strains and are mostly delivered parenterally, limiting the development of an effective mucosal immunity. In this study, we evaluated the potential of intranasal formulations incorporating conserved IAV epitopes, namely the long alpha helix (LAH) of the stalk domain of hemagglutinin and three tandem repeats of the ectodomain of the matrix protein 2 (3M2e), as universal mucosal anti-IAV vaccines in mice and chickens. The IAV epitopes were grafted to nanorings, a novel platform technology for mucosal vaccination formed by the nucleoprotein (N) of the respiratory syncytial virus, in fusion or not with the C-terminal end of the P97 protein (P97c), a recently identified Toll-like receptor 5 agonist. Fusion of LAH to nanorings boosted the generation of LAH-specific systemic and local antibody responses as well as cellular immunity in mice, whereas the carrier effect of nanorings was less pronounced towards 3M2e. Mice vaccinated with chimeric nanorings bearing IAV epitopes in fusion with P97c presented modest LAH- or M2e-specific IgG titers in serum and were unable to generate a mucosal humoral response. In contrast, N-3M2e or N-LAH nanorings admixed with Montanide™ gel (MG) triggered strong specific humoral responses, composed of serum type 1/type 2 IgG and mucosal IgG and IgA, as well as cellular responses dominated by type 1/type 17 cytokine profiles. All mice vaccinated with the [N-3M2e + N-LAH + MG] formulation survived an H1N1 challenge and the combination of both N-3M2e and N-LAH nanorings with MG enhanced the clinical and/or virological protective potential of the preparation in comparison to individual nanorings. Chickens vaccinated parenterally or mucosally with N-LAH and N-3M2e nanorings admixed with Montanide™ adjuvants developed a specific systemic humoral response, which nonetheless failed to confer protection against heterosubtypic challenge with a highly pathogenic H5N8 strain. Thus, while the combination of N-LAH and N-3M2e nanorings with Montanide™ adjuvants shows promise as a universal mucosal anti-IAV vaccine in the mouse model, further experiments have to be conducted to extend its efficacy to poultry.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Chickens
- Cytokines/immunology
- Cytokines/metabolism
- Epitopes/immunology
- Female
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunity, Mucosal/drug effects
- Immunity, Mucosal/immunology
- Immunogenicity, Vaccine/immunology
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/chemistry
- Influenza Vaccines/immunology
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Protective Agents/administration & dosage
- Survival Analysis
- Vaccination/methods
- Mice
Collapse
Affiliation(s)
- Cynthia Calzas
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Molida Mao
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Mathilde Turpaud
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Quentin Viboud
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Joelle Mettier
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Thomas Figueroa
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Pierre Bessière
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Antoine Mangin
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
- Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Laura Sedano
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Pierre-Louis Hervé
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
- Chemistry Department, Université du Québec à Montréal, Montreal, QC, Canada
| | - Romain Volmer
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Mariette F. Ducatez
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, Montreal, QC, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Ronan Le Goffic
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Christophe Chevalier
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
11
|
Noisumdaeng P, Roytrakul T, Prasertsopon J, Pooruk P, Lerdsamran H, Assanasen S, Kitphati R, Auewarakul P, Puthavathana P. T cell mediated immunity against influenza H5N1 nucleoprotein, matrix and hemagglutinin derived epitopes in H5N1 survivors and non-H5N1 subjects. PeerJ 2021; 9:e11021. [PMID: 33854839 PMCID: PMC7955671 DOI: 10.7717/peerj.11021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
Background Protection against the influenza virus by a specific antibody is relatively strain specific; meanwhile broader immunity may be conferred by cell-mediated immune response to the conserved epitopes across influenza virus subtypes. A universal broad-spectrum influenza vaccine which confronts not only seasonal influenza virus, but also avian influenza H5N1 virus is promising. Methods This study determined the specific and cross-reactive T cell responses against the highly pathogenic avian influenza A (H5N1) virus in four survivors and 33 non-H5N1 subjects including 10 H3N2 patients and 23 healthy individuals. Ex vivo IFN-γ ELISpot assay using overlapping peptides spanning the entire nucleoprotein (NP), matrix (M) and hemagglutinin (HA) derived from A/Thailand/1(KAN-1)/2004 (H5N1) virus was employed in adjunct with flow cytometry for determining T cell functions. Microneutralization (microNT) assay was performed to determine the status of previous H5N1 virus infection. Results IFN-γ ELISpot assay demonstrated that survivors nos. 1 and 2 had markedly higher T cell responses against H5N1 NP, M and HA epitopes than survivors nos. 3 and 4; and the magnitude of T cell responses against NP were higher than that of M and HA. Durability of the immunoreactivity persisted for as long as four years after disease onset. Upon stimulation by NP in IFN-γ ELISpot assay, 60% of H3N2 patients and 39% of healthy subjects exhibited a cross-reactive T cell response. The higher frequency and magnitude of responses in H3N2 patients may be due to blood collection at the convalescent phase of the patients. In H5N1 survivors, the effector peptide-specific T cells generated from bulk culture PBMCs by in vitro stimulation displayed a polyfunction by simultaneously producing IFN-γ and TNF-α, together with upregulation of CD107a in recognition of the target cells pulsed with peptide or infected with rVac-NP virus as investigated by flow cytometry. Conclusions This study provides an insight into the better understanding on the homosubtypic and heterosubtypic T cell-mediated immune responses in H5N1 survivors and non-H5N1 subjects. NP is an immunodominant target of cross-recognition owing to its high conservancy. Therefore, the development of vaccine targeting the conserved NP may be a novel strategy for influenza vaccine design.
Collapse
Affiliation(s)
- Pirom Noisumdaeng
- Faculty of Public Health, Thammasat University, Khlong Luang, Pathum Thani, Thailand.,Thammasat University Research Unit in Modern Microbiology and Public Health Genomics, Thammasat University, Khlong Luang, Pathum Thani, Thailand.,Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand
| | - Thaneeya Roytrakul
- National Center for Genetic Engineering and Biotechnology, Khlong Luang, Pathum Thani, Thailand
| | - Jarunee Prasertsopon
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Phisanu Pooruk
- The Government Pharmaceutical Organization, Biological Product Vaccine Production Plant, Kaengkhoi, Saraburi, Thailand
| | - Hatairat Lerdsamran
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Susan Assanasen
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand
| | | | - Prasert Auewarakul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand
| | - Pilaipan Puthavathana
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand.,Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
12
|
Mohn KGI, Brokstad KA, Islam S, Oftung F, Tøndel C, Aarstad HJ, Cox RJ. Early Induction of Cross-Reactive CD8+ T-Cell Responses in Tonsils After Live-Attenuated Influenza Vaccination in Children. J Infect Dis 2021; 221:1528-1537. [PMID: 32255493 PMCID: PMC7137893 DOI: 10.1093/infdis/jiz583] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/10/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Live-attenuated influenza vaccine (LAIV) was licensed for prophylaxis of children 2-17 years old in Europe in 2012 and is administered as a nasal spray. Live-attenuated influenza vaccine induces both mucosal and systemic antibodies and systemic T-cell responses. Tonsils are the lymph nodes serving the upper respiratory tract, acting as both induction and effector site for mucosal immunity. METHODS Here, we have studied the early tonsillar T-cell responses induced in children after LAIV. Thirty-nine children were immunized with trivalent LAIV (containing A/H1N1, A/H3N2, and B viruses) at days 3, 7, and 14 before tonsillectomy. Nonvaccinated controls were included for comparison. Tonsils and peripheral blood (pre- and postvaccination) were collected to study T-cell responses. RESULTS Tonsillar and systemic T-cell responses differed between influenza strains, and both were found against H3N2 and B viruses, whereas only systemic responses were observed against A/H1N1. A significant increase in cross-reactive tonsillar CD8+ T cells recognizing conserved epitopes from a broad range of seasonal and pandemic viruses occurred at day 14. Tonsillar T cells showed significant cytokine responses (Th1, Th2, and granulocyte-macrophage colony-stimulating factor). CONCLUSIONS Our findings support the use of LAIV in children to elicit broadly cross-reactive T cells, which are not induced by traditional inactivated influenza vaccines and may provide protection to novel virus strains.
Collapse
Affiliation(s)
- K G-I Mohn
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| | - K A Brokstad
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - S Islam
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway
| | - F Oftung
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - C Tøndel
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - H J Aarstad
- Department of Otorhinolaryngology/Head and Neck Surgery, Haukeland University Hospital, Bergen, Norway
| | - R J Cox
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
13
|
Law JC, Koh WH, Budylowski P, Lin J, Yue F, Abe KT, Rathod B, Girard M, Li Z, Rini JM, Mubareka S, McGeer A, Chan AK, Gingras AC, Watts TH, Ostrowski MA. Systematic Examination of Antigen-Specific Recall T Cell Responses to SARS-CoV-2 versus Influenza Virus Reveals a Distinct Inflammatory Profile. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:37-50. [PMID: 33208459 PMCID: PMC7750861 DOI: 10.4049/jimmunol.2001067] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
There is a pressing need for an in-depth understanding of immunity to SARS-CoV-2. In this study, we investigated human T cell recall responses to fully glycosylated spike trimer, recombinant N protein, as well as to S, N, M, and E peptide pools in the early convalescent phase and compared them with influenza-specific memory responses from the same donors. All subjects showed SARS-CoV-2-specific T cell responses to at least one Ag. Both SARS-CoV-2-specific and influenza-specific CD4+ T cell responses were predominantly of the central memory phenotype; however SARS-CoV-2-specific CD4+ T cells exhibited a lower IFN-γ to TNF ratio compared with influenza-specific memory responses from the same donors, independent of disease severity. SARS-CoV-2-specific T cells were less multifunctional than influenza-specific T cells, particularly in severe cases, potentially suggesting exhaustion. Most SARS-CoV-2-convalescent subjects also produced IFN-γ in response to seasonal OC43 S protein. We observed granzyme B+/IFN-γ+, CD4+, and CD8+ proliferative responses to peptide pools in most individuals, with CD4+ T cell responses predominating over CD8+ T cell responses. Peripheral T follicular helper (pTfh) responses to S or N strongly correlated with serum neutralization assays as well as receptor binding domain-specific IgA; however, the frequency of pTfh responses to SARS-CoV-2 was lower than the frequency of pTfh responses to influenza virus. Overall, T cell responses to SARS-CoV-2 are robust; however, CD4+ Th1 responses predominate over CD8+ T cell responses, have a more inflammatory profile, and have a weaker pTfh response than the response to influenza virus within the same donors, potentially contributing to COVID-19 disease.
Collapse
Affiliation(s)
- Jaclyn C Law
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Wan Hon Koh
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Patrick Budylowski
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jonah Lin
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - FengYun Yue
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Kento T Abe
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Bhavisha Rathod
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
| | - Melanie Girard
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Zhijie Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - James M Rini
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Samira Mubareka
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Allison McGeer
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Adrienne K Chan
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada; and
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada;
| | - Mario A Ostrowski
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
14
|
Chen Q, Wang L, Xie M, Li X, Recommendations for Influenza, Streptococcus pneumoniae Vaccination in Elderly People in China Writing Group, Geriatric Respiratory Group and Chinese Society of Geriatrics. Recommendations for influenza and Streptococcus pneumoniae vaccination in elderly people in China. Aging Med (Milton) 2020; 3:1-11. [PMID: 32232186 PMCID: PMC7099755 DOI: 10.1002/agm2.12102] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/06/2023] Open
Abstract
Influenza and pneumonia can be prevented by vaccination, but they remain major causes of morbidity and mortality in age-related diseases. In most areas of China, the rates of influenza and pneumococcal vaccination are relatively low and public awareness of vaccination remains insufficient. Thus, it is essential to recommend influenza and Streptococcus pneumoniae vaccination to elderly people in clinical practice. Based on recently published studies and related documents issued by several vaccination authorities, such as the World Health Organization, the National Health and Wellness Committee, the Chinese Center for Disease Control and Prevention, the US Centers for Disease Control and Prevention, and the US Advisory Committee on Immunization Practices, we propose official recommendations for influenza and S pneumoniae vaccination in elderly people in China.
Collapse
Affiliation(s)
- Qiong Chen
- Department of GeriatricsDepartment of Respiratory MedicineXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Lijing Wang
- Department of GeriatricsDepartment of Respiratory MedicineXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Mingxuan Xie
- Department of GeriatricsDepartment of Respiratory MedicineXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Xiaoying Li
- Department of Cardiovascular MedicineChinese PLA General HospitalBeijingChina
| | | |
Collapse
|
15
|
Jang YH, Seong BL. The Quest for a Truly Universal Influenza Vaccine. Front Cell Infect Microbiol 2019; 9:344. [PMID: 31649895 PMCID: PMC6795694 DOI: 10.3389/fcimb.2019.00344] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/24/2019] [Indexed: 12/17/2022] Open
Abstract
There is an unmet public health need for a universal influenza vaccine (UIV) to provide broad and durable protection from influenza virus infections. The identification of broadly protective antibodies and cross-reactive T cells directed to influenza viral targets present a promising prospect for the development of a UIV. Multiple targets for cross-protection have been identified in the stalk and head of hemagglutinin (HA) to develop a UIV. Recently, neuraminidase (NA) has received significant attention as a critical component for increasing the breadth of protection. The HA stalk-based approaches have shown promising results of broader protection in animal studies, and their feasibility in humans are being evaluated in clinical trials. Mucosal immune responses and cross-reactive T cell immunity across influenza A and B viruses intrinsic to live attenuated influenza vaccine (LAIV) have emerged as essential features to be incorporated into a UIV. Complementing the weakness of the stand-alone approaches, prime-boost vaccination combining HA stalk, and LAIV is under clinical evaluation, with the aim to increase the efficacy and broaden the spectrum of protection. Preexisting immunity in humans established by prior exposure to influenza viruses may affect the hierarchy and magnitude of immune responses elicited by an influenza vaccine, limiting the interpretation of preclinical data based on naive animals, necessitating human challenge studies. A consensus is yet to be achieved on the spectrum of protection, efficacy, target population, and duration of protection to define a “universal” vaccine. This review discusses the recent advancements in the development of UIVs, rationales behind cross-protection and vaccine designs, and challenges faced in obtaining balanced protection potency, a wide spectrum of protection, and safety relevant to UIVs.
Collapse
Affiliation(s)
- Yo Han Jang
- Molecular Medicine Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Baik Lin Seong
- Molecular Medicine Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,Vaccine Translational Research Center, Yonsei University, Seoul, South Korea
| |
Collapse
|
16
|
Jansen JM, Gerlach T, Elbahesh H, Rimmelzwaan GF, Saletti G. Influenza virus-specific CD4+ and CD8+ T cell-mediated immunity induced by infection and vaccination. J Clin Virol 2019; 119:44-52. [DOI: 10.1016/j.jcv.2019.08.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/13/2023]
|
17
|
Lee SY, Kang JO, Chang J. Nucleoprotein vaccine induces cross-protective cytotoxic T lymphocytes against both lineages of influenza B virus. Clin Exp Vaccine Res 2019; 8:54-63. [PMID: 30775351 PMCID: PMC6369129 DOI: 10.7774/cevr.2019.8.1.54] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/14/2019] [Indexed: 12/31/2022] Open
Abstract
Purpose The influenza B virus diverges into two antigenically distinct lineages: B/Yamagata and B/Victoria. Influenza B is the dominant circulating virus during some influenza seasons, and recent data demonstrated that influenza A and B infection similarly cause severe clinical symptoms in hospitalized patients. Nucleoprotein (NP) is a good target for a universal influenza vaccine. This study investigated whether NP epitope variation within two lineages affects the dominant cytotoxic T lymphocyte (CTL) responses induced by vaccination and the resultant protective immunity. Materials and Methods The NP of B/Yamagata/16/1988, the representative strain of the Yamagata lineage, includes a dominant CTL epitope, FSPIRITFL, while B/Shangdong/7/1997 from the Victoria lineage has one amino acid difference in this sequence, FSPIRVTFL. Two recombinant replication-deficient adenovirus (rAd)-vectored vaccines expressing either NP were prepared (rAd/B-NP(I) and rAd/B-NP(V), respectively) and administered to BALB/c mice intranasally. To examine the efficacy of vaccination, antibody responses, CTL responses, and morbidity/mortality after challenge were measured. Results Both vaccines induce similar antibody and CD8 T-cell responses cross-reacting to both epitopes, and also confer cross-protection against both lineages regardless of amino acid difference. Conclusion The rAd-vectored vaccine expressing the NP could be developed as universal influenza B vaccine which provides broader protection.
Collapse
Affiliation(s)
- So-Young Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Jung-Ok Kang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| |
Collapse
|
18
|
Yamayoshi S, Kawaoka Y. Current and future influenza vaccines. Nat Med 2019; 25:212-220. [PMID: 30692696 DOI: 10.1038/s41591-018-0340-z] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/19/2018] [Indexed: 11/09/2022]
Abstract
Although antiviral drugs and vaccines have reduced the economic and healthcare burdens of influenza, influenza epidemics continue to take a toll. Over the past decade, research on influenza viruses has revealed a potential path to improvement. The clues have come from accumulated discoveries from basic and clinical studies. Now, virus surveillance allows researchers to monitor influenza virus epidemic trends and to accumulate virus sequences in public databases, which leads to better selection of candidate viruses for vaccines and early detection of drug-resistant viruses. Here we provide an overview of current vaccine options and describe efforts directed toward the development of next-generation vaccines. Finally, we propose a plan for the development of an optimal influenza vaccine.
Collapse
Affiliation(s)
- Seiya Yamayoshi
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan. .,Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan. .,Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin Madison, Madison, WI, USA.
| |
Collapse
|
19
|
Broadened immunity against influenza by vaccination with computationally designed influenza virus N1 neuraminidase constructs. NPJ Vaccines 2018; 3:55. [PMID: 30510776 PMCID: PMC6265323 DOI: 10.1038/s41541-018-0093-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022] Open
Abstract
Split inactivated influenza vaccines remain one of the primary preventative strategies against severe influenza disease in the population. However, current vaccines are only effective against a limited number of matched strains. The need for broadly protective vaccines is acute due to the high mutational rate of influenza viruses and multiple strain variants in circulation at any one time. The neuraminidase (NA) glycoprotein expressed on the influenza virion surface has recently regained recognition as a valuable vaccine candidate. We sought to broaden the protection provided by NA within the N1 subtype by computationally engineering consensus NA sequences. Three NA antigens (NA5200, NA7900, NA9100) were designed based on sequence clusters encompassing three major groupings of NA sequence space; (i) H1N1 2009 pandemic and Swine H1N1, (ii) historical seasonal H1N1 and (iii) H1N1 viruses ranging from 1933 till current times. Recombinant NA proteins were produced as a vaccine and used in a mouse challenge model. The design of the protein dictated the protection provided against the challenge strains. NA5200 protected against H1N1 pdm09, a Swine isolate from 1998 and NIBRG-14 (H5N1). NA7900 protected against all seasonal H1N1 viruses tested, and NA9100 showed the broadest range of protection covering all N1 viruses tested. By passive transfer studies and serological assays, the protection provided by the cluster-based consensus (CBC) designs correlated to antibodies capable of mediating NA inhibition. Importantly, sera raised to the consensus NAs displayed a broader pattern of reactivity and protection than naturally occurring NAs, potentially supporting a predictive approach to antigen design. The high variability of the influenza virus — arising from its high mutation rate and wide range of strains — limits the effectiveness of influenza vaccines unless they induce a broad immune response, a difficult task when relying on natural viral antigens. Here, Xavier Saelens, Thorsten Vogel, Ray Oomen and colleagues applied a ‘cluster-based’ consensus computational approach to design three consensus sequences of the viral protein neuroaminidase (NA) subtype 1 that induce broadly protective immune responses in vaccinated mice. NA9100, a consensus NA sequence based on H1N1 virus strains collected from 1933 to today, was protective against all N1 viruses tested. By using a computational method to integrate multiple sequences of viral proteins into one consensus protein, the researchers provide a strategy that can be applied to develop broadly protective vaccine formulations for influenza virus.
Collapse
|
20
|
Mohn KGI, Zhou F. Clinical Expectations for Better Influenza Virus Vaccines-Perspectives from the Young Investigators' Point of View. Vaccines (Basel) 2018; 6:E32. [PMID: 29861454 PMCID: PMC6027204 DOI: 10.3390/vaccines6020032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/20/2023] Open
Abstract
The influenza virus is one of a few viruses that is capable of rendering an otherwise healthy person acutly bedridden for several days. This impressive knock-out effect, without prodromal symptoms, challenges our immune system. The influenza virus undergoes continuous mutations, escaping our pre-existing immunity and causing epidemics, and its segmented genome is subject to reassortment, resulting in novel viruses with pandemic potential. The personal and socieoeconomic burden from influenza is high. Vaccination is the most cost-effective countermeasure, with several vaccines that are available. The current limitations in vaccine effectivness, combined with the need for yearly updating of vaccine strains, is a driving force for research into developing new and improved influenza vaccines. The lack of public concern about influenza severity, and misleading information concerning vaccine safety contribute to low vaccination coverage even in high-risk groups. The success of future influeza vaccines will depend on an increased public awarness of the disease, and hence, the need for vaccination-aided through improved rapid diagnositics. The vaccines must be safe and broadly acting, with new, measurable correlates of protection and robust post-marketing safety studies, to improve the confidence in influenza vaccines.
Collapse
Affiliation(s)
- Kristin G-I Mohn
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
- Emergency Care clinic, Haukeland University Hospital, Bergen 5021, Norway.
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
| |
Collapse
|
21
|
Sebastian S, Lambe T. Clinical Advances in Viral-Vectored Influenza Vaccines. Vaccines (Basel) 2018; 6:E29. [PMID: 29794983 PMCID: PMC6027524 DOI: 10.3390/vaccines6020029] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 12/27/2022] Open
Abstract
Influenza-virus-mediated disease can be associated with high levels of morbidity and mortality, particularly in younger children and older adults. Vaccination is the primary intervention used to curb influenza virus infection, and the WHO recommends immunization for at-risk individuals to mitigate disease. Unfortunately, influenza vaccine composition needs to be updated annually due to antigenic shift and drift in the viral immunogen hemagglutinin (HA). There are a number of alternate vaccination strategies in current development which may circumvent the need for annual re-vaccination, including new platform technologies such as viral-vectored vaccines. We discuss the different vectored vaccines that have been or are currently in clinical trials, with a forward-looking focus on immunogens that may be protective against seasonal and pandemic influenza infection, in the context of viral-vectored vaccines. We also discuss future perspectives and limitations in the field that will need to be addressed before new vaccines can significantly impact disease levels.
Collapse
Affiliation(s)
- Sarah Sebastian
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 DQ, UK.
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 DQ, UK.
| |
Collapse
|
22
|
Qi M, Zhang XE, Sun X, Zhang X, Yao Y, Liu S, Chen Z, Li W, Zhang Z, Chen J, Cui Z. Intranasal Nanovaccine Confers Homo- and Hetero-Subtypic Influenza Protection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703207. [PMID: 29430819 DOI: 10.1002/smll.201703207] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 01/03/2018] [Indexed: 05/21/2023]
Abstract
Cross-protective and non-invasively administered vaccines are attractive and highly desired for the control of influenza. Self-assembling nanotechnology provides an opportunity for the development of vaccines with superior performance. In this study, an intranasal nanovaccine is developed targeting the conserved ectodomain of influenza matrix protein 2(M2e). 3-sequential repeats of M2e (3M2e) is presented on the self-assembling recombinant human heavy chain ferritin (rHF) cage to form the 3M2e-rHF nanoparticle. Intranasal vaccination with 3M2e-rHF nanoparticles in the absence of an adjuvant induces robust immune responses, including high titers of sera M2e-specific IgG antibodies, T-cell immune responses, and mucosal secretory-IgA antibodies in mice. The 3M2e-rHF nanoparticles also confer complete protection against a lethal infection of homo-subtypic H1N1 and hetero-subtypic H9N2 virus. An analysis of the mechanism of protection underlying the intranasal immunization with the 3M2e-rHF nanoparticle indicates that M2e-specific mucosal secretory-IgA and T-cell immune responses may play critical roles in the prevention of infection. The results suggest that the 3M2e-rHF nanoparticle is a promising, needle-free, intranasally administered, cross-protective influenza vaccine. The use of self-assembling nanovaccines could be an ideal strategy for developing vaccines with characteristics such as high immunogenicity, cross-protection, and convenient administration, as well as being economical and suitable for large-scale production.
Collapse
Affiliation(s)
- Mi Qi
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian-En Zhang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xianxun Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yanfeng Yao
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science and Technology, Wuhan, 430208, China
| | - Siling Liu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ze Chen
- College of Life Sciences, Hunan Normal University, Changsha, 410013, China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhiping Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jianjun Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| |
Collapse
|
23
|
Muraro E, Merlo A, Martorelli D, Cangemi M, Dalla Santa S, Dolcetti R, Rosato A. Fighting Viral Infections and Virus-Driven Tumors with Cytotoxic CD4 + T Cells. Front Immunol 2017; 8:197. [PMID: 28289418 PMCID: PMC5327441 DOI: 10.3389/fimmu.2017.00197] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/09/2017] [Indexed: 12/18/2022] Open
Abstract
CD4+ T cells have been and are still largely regarded as the orchestrators of immune responses, being able to differentiate into distinct T helper cell populations based on differentiation signals, transcription factor expression, cytokine secretion, and specific functions. Nonetheless, a growing body of evidence indicates that CD4+ T cells can also exert a direct effector activity, which depends on intrinsic cytotoxic properties acquired and carried out along with the evolution of several pathogenic infections. The relevant role of CD4+ T cell lytic features in the control of such infectious conditions also leads to their exploitation as a new immunotherapeutic approach. This review aims at summarizing currently available data about functional and therapeutic relevance of cytotoxic CD4+ T cells in the context of viral infections and virus-driven tumors.
Collapse
Affiliation(s)
- Elena Muraro
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | - Anna Merlo
- Department of Immunology and Blood Transfusions, San Bortolo Hospital, Vicenza, Italy
| | - Debora Martorelli
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | - Michela Cangemi
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | | | - Riccardo Dolcetti
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
- Translational Research Institute, University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Antonio Rosato
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padova, Padova, Italy
| |
Collapse
|
24
|
Increased Protein Degradation Improves Influenza Virus Nucleoprotein-Specific CD8+ T Cell Activation In Vitro but Not in C57BL/6 Mice. J Virol 2016; 90:10209-10219. [PMID: 27581985 DOI: 10.1128/jvi.01633-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/25/2016] [Indexed: 11/20/2022] Open
Abstract
Due to antigenic drift of influenza viruses, seasonal influenza vaccines need to be updated annually. These vaccines are based on predictions of strains likely to circulate in the next season. However, vaccine efficacy is greatly reduced in the case of a mismatch between circulating and vaccine strains. Furthermore, novel antigenically distinct influenza viruses are introduced into the human population from animal reservoirs occasionally and may cause pandemic outbreaks. To dampen the impact of seasonal and pandemic influenza, vaccines that induce broadly protective and long-lasting immunity are preferred. Because influenza virus-specific CD8+ T cells are directed mainly against relatively conserved internal proteins, like nucleoprotein (NP), they are highly cross-reactive and afford protection against infection with antigenically distinct influenza virus strains, so-called heterosubtypic immunity. Here, we used modified vaccinia virus Ankara (MVA) as a vaccine vector for the induction of influenza virus NP-specific CD8+ T cells. To optimize the induction of CD8+ T cell responses, we made several modifications to NP, aiming at retaining the protein in the cytosol or targeting it to the proteasome. We hypothesized that these strategies would increase antigen processing and presentation and thus improve the induction of CD8+ T cell responses. We showed that NP with increased degradation rates improved CD8+ T cell activation in vitro if the amount of antigen was limited or if CD8+ T cells were of low functional avidity. However, after immunization of C57BL/6 mice, no differences were detected between modified NP and wild-type NP (NPwt), since NPwt already induced optimal CD8+ T cell responses. IMPORTANCE Due to the continuous antigenic drift of seasonal influenza viruses and the threat of a novel pandemic, there is a great need for the development of novel influenza vaccines that offer broadly protective immunity against multiple subtypes. CD8+ T cells can provide immunity against multiple subtypes of influenza viruses by the recognition of relatively conserved internal antigens. In this study, we aimed at optimizing the CD8+ T cell response to influenza A virus by making modifications to influenza A virus nucleoprotein (NP) expressed from the modified vaccinia virus Ankara (MVA) vaccine vector. These modifications resulted in increased antigen degradation, thereby producing elevated levels of peptides that can be presented on major histocompatibility complex (MHC) class I molecules to CD8+ T cells. Although we were unable to increase the NP-specific immune response in the mouse strain used, this approach may have benefits for vaccine development using less-immunogenic proteins.
Collapse
|
25
|
Kolpe A, Schepens B, Fiers W, Saelens X. M2-based influenza vaccines: recent advances and clinical potential. Expert Rev Vaccines 2016; 16:123-136. [DOI: 10.1080/14760584.2017.1240041] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Annasaheb Kolpe
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bert Schepens
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Walter Fiers
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
26
|
DiPiazza A, Richards K, Batarse F, Lockard L, Zeng H, García-Sastre A, Albrecht RA, Sant AJ. Flow Cytometric and Cytokine ELISpot Approaches To Characterize the Cell-Mediated Immune Response in Ferrets following Influenza Virus Infection. J Virol 2016; 90:7991-8004. [PMID: 27356897 PMCID: PMC4988159 DOI: 10.1128/jvi.01001-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/18/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Influenza virus infections represent a significant socioeconomic and public health burden worldwide. Although ferrets are considered by many to be ideal for modeling human responses to influenza infection and vaccination, efforts to understand the cellular immune response have been severely hampered by a paucity of standardized procedures and reagents. In this study, we developed flow cytometric and T cell enzyme-linked immunosorbent spot (ELISpot) approaches to characterize the leukocyte composition and antigen-specific T cell response within key lymphoid tissues following influenza virus infection in ferrets. Through a newly designed and implemented set of serological reagents, we used multiparameter flow cytometry to directly quantify the frequency of CD4(+) and CD8(+) T cells, Ig(+) B cells, CD11b(+) myeloid-derived cells, and major histocompatibility complex (MHC) class II-positive antigen-presenting cells (APCs) both prior to and after intranasal infection with A/California/04/09 (H1N1). We found that the leukocyte composition was altered at 10 days postinfection, with notable gains in the frequency of T cells and myeloid cells within the draining lymph node. Furthermore, these studies revealed that the antigen specificity of influenza virus-reactive CD4 and CD8 T cells was very broad, with recognition of the viral HA, NA, M1, NS1, and NP proteins, and that total reactivity to influenza virus postinfection represented approximately 0.1% of the circulating peripheral blood mononuclear cells (PBMC). Finally, we observed distinct patterns of reactivity between individual animals, suggesting heterogeneity at the MHC locus in ferrets within commercial populations, a finding of considerable interest in efforts to move the ferret model forward for influenza vaccine and challenge studies. IMPORTANCE Ferrets are an ideal animal model to study transmission, diseases, and vaccine efficacies of respiratory viruses because of their close anatomical and physiological resemblances to humans. However, a lack of reagents has limited our understanding of the cell-mediated immune response following infection and vaccination. In this study, we used cross-reactive and ferret-specific antibodies to study the leukocyte composition and antigen-specific CD4 and CD8 T cell responses following influenza A/California/04/09 (H1N1) virus infection. These studies revealed strikingly distinct patterns of reactivity between CD4 and CD8 T cells, which were overlaid with differences in protein-specific responses between individual animals. Our results provide a first, in-depth look at the T cell repertoire in response to influenza infection and suggest that there is considerable heterogeneity at the MHC locus, which is akin to that in humans and an area of intense research interest.
Collapse
Affiliation(s)
- Anthony DiPiazza
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Katherine Richards
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Frances Batarse
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Laura Lockard
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Hui Zeng
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mt. Sinai, New York, New York, USA Global Health and Emerging Pathogens Institute at Icahn School of Medicine, New York, New York, USA Department of Medicine, Icahn School of Medicine at Mt. Sinai, New York, New York, USA
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mt. Sinai, New York, New York, USA Global Health and Emerging Pathogens Institute at Icahn School of Medicine, New York, New York, USA
| | - Andrea J Sant
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
27
|
Self-Amplifying mRNA Vaccines Expressing Multiple Conserved Influenza Antigens Confer Protection against Homologous and Heterosubtypic Viral Challenge. PLoS One 2016; 11:e0161193. [PMID: 27525409 PMCID: PMC4985159 DOI: 10.1371/journal.pone.0161193] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/01/2016] [Indexed: 11/19/2022] Open
Abstract
Current hemagglutinin (HA)-based seasonal influenza vaccines induce vaccine strain-specific neutralizing antibodies that usually fail to provide protection against mismatched circulating viruses. Inclusion in the vaccine of highly conserved internal proteins such as the nucleoprotein (NP) and the matrix protein 1 (M1) was shown previously to increase vaccine efficacy by eliciting cross-reactive T-cells. However, appropriate delivery systems are required for efficient priming of T-cell responses. In this study, we demonstrated that administration of novel self-amplifying mRNA (SAM®) vectors expressing influenza NP (SAM(NP)), M1 (SAM(M1)), and NP and M1 (SAM(M1-NP)) delivered with lipid nanoparticles (LNP) induced robust polyfunctional CD4 T helper 1 cells, while NP-containing SAM also induced cytotoxic CD8 T cells. Robust expansions of central memory (TCM) and effector memory (TEM) CD4 and CD8 T cells were also measured. An enhanced recruitment of NP-specific cytotoxic CD8 T cells was observed in the lungs of SAM(NP)-immunized mice after influenza infection that paralleled with reduced lung viral titers and pathology, and increased survival after homologous and heterosubtypic influenza challenge. Finally, we demonstrated for the first time that the co-administration of RNA (SAM(M1-NP)) and protein (monovalent inactivated influenza vaccine (MIIV)) was feasible, induced simultaneously NP-, M1- and HA-specific T cells and HA-specific neutralizing antibodies, and enhanced MIIV efficacy against a heterologous challenge. In conclusion, systemic administration of SAM vectors expressing conserved internal influenza antigens induced protective immune responses in mice, supporting the SAM® platform as another promising strategy for the development of broad-spectrum universal influenza vaccines.
Collapse
|
28
|
Dash SK, Kumar M, Kataria JM, Nagarajan S, Tosh C, Murugkar HV, Kulkarni DD. Partial heterologous protection by low pathogenic H9N2 virus against natural H9N2-PB1 gene reassortant highly pathogenic H5N1 virus in chickens. Microb Pathog 2016; 95:157-165. [DOI: 10.1016/j.micpath.2016.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 04/01/2016] [Accepted: 04/01/2016] [Indexed: 11/30/2022]
|
29
|
Sridhar S. Heterosubtypic T-Cell Immunity to Influenza in Humans: Challenges for Universal T-Cell Influenza Vaccines. Front Immunol 2016; 7:195. [PMID: 27242800 PMCID: PMC4871858 DOI: 10.3389/fimmu.2016.00195] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/03/2016] [Indexed: 11/25/2022] Open
Abstract
Influenza A virus (IAV) remains a significant global health issue causing annual epidemics, pandemics, and sporadic human infections with highly pathogenic avian or swine influenza viruses. Current inactivated and live vaccines are the mainstay of the public health response to influenza, although vaccine efficacy is lower against antigenically distinct viral strains. The first pandemic of the twenty-first century underlined the urgent need to develop new vaccines capable of protecting against a broad range of influenza strains. Such “universal” influenza vaccines are based on the idea of heterosubtypic immunity, wherein immune responses to epitopes conserved across IAV strains can confer protection against subsequent infection and disease. T-cells recognizing conserved antigens are a key contributor in reducing viral load and limiting disease severity during heterosubtypic infection in animal models. Recent studies undertaken during the 2009 H1N1 pandemic provided key insights into the role of cross-reactive T-cells in mediating heterosubtypic protection in humans. This review focuses on human influenza to discuss the epidemiological observations that underpin cross-protective immunity, the role of T-cells as key players in mediating heterosubtypic immunity including recent data from natural history cohort studies and the ongoing clinical development of T-cell-inducing universal influenza vaccines. The challenges and knowledge gaps for developing vaccines to generate long-lived protective T-cell responses is discussed.
Collapse
|
30
|
Castilla J, Navascués A, Fernández-Alonso M, Reina G, Pozo F, Casado I, Guevara M, Martínez-Baz I, Barricarte A, Ezpeleta C. Effectiveness of subunit influenza vaccination in the 2014-2015 season and residual effect of split vaccination in previous seasons. Vaccine 2016; 34:1350-7. [PMID: 26854911 DOI: 10.1016/j.vaccine.2016.01.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 01/08/2016] [Accepted: 01/26/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND In Navarra, Spain, subunit vaccine was first used in the 2014-2015 season, whereas trivalent split-virion influenza vaccines had been used in previous seasons. We estimate the effectiveness of the subunit vaccine in the current season and split vaccine in the two previous seasons against laboratory-confirmed influenza in the 2014-2015 season. METHODS Patients with influenza-like illness hospitalized or attended by sentinel general practitioners were swabbed for influenza testing. The previous and current vaccine status of laboratory-confirmed cases was compared to test-negative controls. RESULTS Among 1213 patients tested, 619 (51%) were confirmed for influenza virus: 52% influenza A(H3N2), 46% influenza B, and 2% A(H1N1)pdm09. The overall effectiveness for subunit vaccination in the current season was 19% (95% confidence interval [CI]: -13 to 42), 2% (95%CI: -47 to 35) against influenza A(H3N2) and 32% (95%CI: -4 to 56) against influenza B. The effectiveness against any influenza was 67% (95%CI: 17-87) for 2012-2013 and 2013-2014 vaccination only, 42% (95%CI: -31 to 74) for 2014-2015 vaccination only, and 38% (95%CI: 8-58) for vaccination in the 2012-2013, 2013-2014 and 2014-2015 seasons. The same estimates against influenza A(H3N2) were 47% (95%CI: -60 to 82), -54% (95%CI: -274 to 37) and 28% (95%CI: -17 to 56), and against influenza B were 82% (95%CI: 19-96), 93% (95%CI: 45-99) and 43% (95%CI: 5-66), respectively. CONCLUSION These results suggest a considerable residual protection of split vaccination in previous seasons, low overall effectiveness of current season subunit vaccination, and possible interference between current subunit and previous split vaccines.
Collapse
Affiliation(s)
- Jesús Castilla
- Instituto de Salud Pública de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Spain.
| | - Ana Navascués
- Complejo Hospitalario de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain
| | - Mirian Fernández-Alonso
- Clínica Universidad de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain
| | - Gabriel Reina
- Clínica Universidad de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain
| | - Francisco Pozo
- Centro Nacional de Microbiología (WHO National Influenza Centre-Madrid), Instituto de Salud Carlos III, Majadahonda, Spain
| | - Itziar Casado
- Instituto de Salud Pública de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Marcela Guevara
- Instituto de Salud Pública de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Iván Martínez-Baz
- Instituto de Salud Pública de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Aurelio Barricarte
- Instituto de Salud Pública de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Carmen Ezpeleta
- Complejo Hospitalario de Navarra, IdiSNA-Navarra Institute for Health Research, Pamplona, Spain
| | | | | |
Collapse
|
31
|
Lee YN, Kim MC, Lee YT, Kim YJ, Kang SM. Mechanisms of Cross-protection by Influenza Virus M2-based Vaccines. Immune Netw 2015; 15:213-21. [PMID: 26557805 PMCID: PMC4637342 DOI: 10.4110/in.2015.15.5.213] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 08/25/2015] [Accepted: 09/10/2015] [Indexed: 01/06/2023] Open
Abstract
Current influenza virus vaccines are based on strain-specific surface glycoprotein hemagglutinin (HA) antigens and effective only when the predicted vaccine strains and circulating viruses are well-matched. The current strategy of influenza vaccination does not prevent the pandemic outbreaks and protection efficacy is reduced or ineffective if mutant strains emerge. It is of high priority to develop effective vaccines and vaccination strategies conferring a broad range of cross protection. The extracellular domain of M2 (M2e) is highly conserved among human influenza A viruses and has been utilized to develop new vaccines inducing cross protection against different subtypes of influenza A virus. However, immune mechanisms of cross protection by M2e-based vaccines still remain to be fully elucidated. Here, we review immune correlates and mechanisms conferring cross protection by M2e-based vaccines. Molecular and cellular immune components that are known to be involved in M2 immune-mediated protection include antibodies, B cells, T cells, alveolar macrophages, Fc receptors, complements, and natural killer cells. Better understanding of protective mechanisms by immune responses induced by M2e vaccination will help facilitate development of broadly cross protective vaccines against influenza A virus.
Collapse
Affiliation(s)
- Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA. ; Animal and Plant Quarantine Agency, Anyang 14089, Korea
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
32
|
van de Sandt CE, Bodewes R, Rimmelzwaan GF, de Vries RD. Influenza B viruses: not to be discounted. Future Microbiol 2015; 10:1447-65. [PMID: 26357957 DOI: 10.2217/fmb.15.65] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In contrast to influenza A viruses, which have been investigated extensively, influenza B viruses have attracted relatively little attention. However, influenza B viruses are an important cause of morbidity and mortality in the human population and full understanding of their biological and epidemiological properties is imperative to better control this important pathogen. However, some of its characteristics are still elusive and warrant investigation. Here, we review evolution, epidemiology, pathogenesis and immunity and identify gaps in our knowledge of influenza B viruses. The divergence of two antigenically distinct influenza B viruses is highlighted. The co-circulation of viruses of these two lineages necessitated the development of quadrivalent influenza vaccines, which is discussed in addition to possibilities to develop universal vaccination strategies.
Collapse
Affiliation(s)
- Carolien E van de Sandt
- Department of Viroscience, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Rogier Bodewes
- Department of Viroscience, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,ViroClinics Biosciences BV, Rotterdam Science Tower, Marconistraat 16, 3029 AK Rotterdam, The Netherlands
| | - Rory D de Vries
- Department of Viroscience, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Using epidemics to map H3 equine influenza virus determinants of antigenicity. Virology 2015; 481:187-98. [DOI: 10.1016/j.virol.2015.02.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/08/2014] [Accepted: 02/14/2015] [Indexed: 01/25/2023]
|
34
|
van de Sandt CE, Dou Y, Vogelzang-van Trierum SE, Westgeest KB, Pronk MR, Osterhaus ADME, Fouchier RAM, Rimmelzwaan GF, Hillaire MLB. Influenza B virus-specific CD8+ T-lymphocytes strongly cross-react with viruses of the opposing influenza B lineage. J Gen Virol 2015; 96:2061-2073. [PMID: 25900135 DOI: 10.1099/vir.0.000156] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza B viruses fall in two antigenically distinct lineages (B/Victoria/2/1987 and B/Yamagata/16/1988 lineage) that co-circulate with influenza A viruses of the H3N2 and H1N1 subtypes during seasonal epidemics. Infections with influenza B viruses contribute considerably to morbidity and mortality in the human population. Influenza B virus neutralizing antibodies, elicited by natural infections or vaccination, poorly cross-react with viruses of the opposing influenza B lineage. Therefore, there is an increased interest in identifying other correlates of protection which could aid the development of broadly protective vaccines. blast analysis revealed high sequence identity of all viral proteins. With two online epitope prediction algorithms, putative conserved epitopes relevant for study subjects used in the present study were predicted. The cross-reactivity of influenza B virus-specific polyclonal CD8+ cytotoxic T-lymphocyte (CTL) populations obtained from HLA-typed healthy study subjects, with intra-lineage drift variants and viruses of the opposing lineage, was determined by assessing their in vitro IFN-γ response and lytic activity. Here, we show for the first time, to the best of our knowledge, that CTLs directed to viruses of the B/Victoria/2/1987 lineage cross-react with viruses of the B/Yamagata/16/1988 lineage and vice versa.
Collapse
Affiliation(s)
| | - YingYing Dou
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | - Kim B Westgeest
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Mark R Pronk
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Albert D M E Osterhaus
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands.,ViroClinics Biosciences BV, Rotterdam, The Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Guus F Rimmelzwaan
- ViroClinics Biosciences BV, Rotterdam, The Netherlands.,Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
35
|
Talbot HK, Nian H, Zhu Y, Chen Q, Williams JV, Griffin MR. Clinical effectiveness of split-virion versus subunit trivalent influenza vaccines in older adults. Clin Infect Dis 2015; 60:1170-5. [PMID: 25697739 PMCID: PMC4447778 DOI: 10.1093/cid/civ019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/24/2014] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Inactivated influenza vaccines are manufactured using either split-virion or subunit methods. These 2 methods produce similar hemagglutinin antibody responses, but different cellular immune responses. METHODS We compared the effectiveness of split-virion influenza vaccines to that of subunit influenza vaccines using prospectively collected data from adults aged ≥50 years who sought care for acute respiratory illness during 3 influenza seasons: 2008-2009, 2010-2011, and 2011-2012 using a case-positive, control test-negative study design. RESULTS Complete data were available for 539 participants, of whom 68 (12.6%) had influenza detected. Influenza-infected patients were younger (P<.001), were more likely to have received no vaccine or the subunit influenza vaccine than the split-virion vaccine (P<.001), and more likely to have sought care in either the emergency department or the acute care clinic than the hospital (P=.001). Split-virion vaccine effectiveness was 77.8% (95% confidence interval [CI], 58.5%-90.3%) compared with subunit vaccine effectiveness of 44.2% (95% CI, -11.8% to 70.9%), giving a difference in vaccine effectiveness of 33.5% (95% CI, 6.9%-86.7%). CONCLUSIONS Studies need to be done to further explore if there are differences in clinical effectiveness in older adults for the 2 vaccine manufacturing methods.
Collapse
Affiliation(s)
- H Keipp Talbot
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hui Nian
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yuwei Zhu
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qingxia Chen
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | |
Collapse
|
36
|
Deng L, Cho KJ, Fiers W, Saelens X. M2e-Based Universal Influenza A Vaccines. Vaccines (Basel) 2015; 3:105-36. [PMID: 26344949 PMCID: PMC4494237 DOI: 10.3390/vaccines3010105] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 12/23/2014] [Accepted: 01/30/2015] [Indexed: 12/13/2022] Open
Abstract
The successful isolation of a human influenza virus in 1933 was soon followed by the first attempts to develop an influenza vaccine. Nowadays, vaccination is still the most effective method to prevent human influenza disease. However, licensed influenza vaccines offer protection against antigenically matching viruses, and the composition of these vaccines needs to be updated nearly every year. Vaccines that target conserved epitopes of influenza viruses would in principle not require such updating and would probably have a considerable positive impact on global human health in case of a pandemic outbreak. The extracellular domain of Matrix 2 (M2e) protein is an evolutionarily conserved region in influenza A viruses and a promising epitope for designing a universal influenza vaccine. Here we review the seminal and recent studies that focused on M2e as a vaccine antigen. We address the mechanism of action and the clinical development of M2e-vaccines. Finally, we try to foresee how M2e-based vaccines could be implemented clinically in the future.
Collapse
Affiliation(s)
- Lei Deng
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Ki Joon Cho
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Walter Fiers
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Xavier Saelens
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| |
Collapse
|
37
|
Feng L, Yang P, Zhang T, Yang J, Fu C, Qin Y, Zhang Y, Ma C, Liu Z, Wang Q, Zhao G, Yu H. Technical guidelines for the application of seasonal influenza vaccine in China (2014-2015). Hum Vaccin Immunother 2015; 11:2077-101. [PMID: 26042462 PMCID: PMC4635867 DOI: 10.1080/21645515.2015.1027470] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/05/2015] [Indexed: 10/23/2022] Open
Abstract
Influenza, caused by the influenza virus, is a respiratory infectious disease that can severely affect human health. Influenza viruses undergo frequent antigenic changes, thus could spread quickly. Influenza causes seasonal epidemics and outbreaks in public gatherings such as schools, kindergartens, and nursing homes. Certain populations are at risk for severe illness from influenza, including pregnant women, young children, the elderly, and people in any ages with certain chronic diseases.
Collapse
Affiliation(s)
- Luzhao Feng
- Key Laboratory of Surveillance and Early-warning on Infectious Disease; Division of Infectious Disease; Chinese Center for Disease Control and Prevention; Beijing, China
| | - Peng Yang
- Beijing Center for Disease Control and Prevention; Beijing, China
| | - Tao Zhang
- School of Public Health; Fudan University; Shanghai, China
| | - Juan Yang
- Key Laboratory of Surveillance and Early-warning on Infectious Disease; Division of Infectious Disease; Chinese Center for Disease Control and Prevention; Beijing, China
| | - Chuanxi Fu
- Guangzhou Center for Disease Control and Prevention; Guangzhou, China
| | - Ying Qin
- Key Laboratory of Surveillance and Early-warning on Infectious Disease; Division of Infectious Disease; Chinese Center for Disease Control and Prevention; Beijing, China
| | - Yi Zhang
- Beijing Center for Disease Control and Prevention; Beijing, China
| | - Chunna Ma
- Beijing Center for Disease Control and Prevention; Beijing, China
| | - Zhaoqiu Liu
- Hua Xin Hospital; First Hospital of Tsinghua University; Beijing, China
| | - Quanyi Wang
- Beijing Center for Disease Control and Prevention; Beijing, China
| | - Genming Zhao
- School of Public Health; Fudan University; Shanghai, China
| | - Hongjie Yu
- Key Laboratory of Surveillance and Early-warning on Infectious Disease; Division of Infectious Disease; Chinese Center for Disease Control and Prevention; Beijing, China
| |
Collapse
|
38
|
Bengs S, Marttila J, Susi P, Ilonen J. Elicitation of T-cell responses by structural and non-structural proteins of coxsackievirus B4. J Gen Virol 2014; 96:322-330. [PMID: 25381056 DOI: 10.1099/vir.0.069062-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Coxsackievirus B4 (CV-B4) belongs to the genus Enterovirus within the family Picornaviridae. To investigate target proteins recognized by T-cells in human enterovirus B infections, virus-encoded structural [VP0 (VP4 and VP2), VP1, VP3] and non-structural (2A, 2B, 2C, 3C and 3D) proteins were expressed and purified in Escherichia coli. Peripheral blood of 19 healthy adult donors was used to create enterovirus-specific T-cell lines by repeated stimulation with CV-B4 cell lysate antigen. T-cell lines responded in individual patterns, and responses to all purified proteins were observed. The most often recognized enteroviral protein was VP0, which is the fusion between the most conserved structural proteins, VP4 and VP2. T-cell responses to VP0 were detected in 15 of the 19 (79 %) donor lines. Non-structural 2C protein was recognized in 11 of the 19 (58 %) lines, and 11 of the 19 (58 %) lines also had a response to 3D protein. Furthermore, responses to other non-structural proteins (2A, 2B and 3C) were also detected. T-cell responses did not correlate clearly to the individual HLA-DR-DQ phenotype or the history of past coxsackie B virus infections of the donors.
Collapse
Affiliation(s)
- Suvi Bengs
- Department of Virology, University of Turku, Turku, Finland.,Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Jane Marttila
- Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Petri Susi
- Biomaterials and Diagnostics Group, Turku University of Applied Sciences, Turku, Finland.,Department of Virology, University of Turku, Turku, Finland
| | - Jorma Ilonen
- Department of Clinical Microbiology, University of Eastern Finland, Kuopio, Finland.,Immunogenetics Laboratory, University of Turku, Turku, Finland
| |
Collapse
|
39
|
Lee YN, Lee YT, Kim MC, Hwang HS, Lee JS, Kim KH, Kang SM. Fc receptor is not required for inducing antibodies but plays a critical role in conferring protection after influenza M2 vaccination. Immunology 2014; 143:300-9. [PMID: 24773389 DOI: 10.1111/imm.12310] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 12/29/2022] Open
Abstract
The ectodomain of matrix protein 2 (M2e) of influenza virus is considered a rational target for a universal influenza A vaccine. To better understand M2e immune-mediated protection, Fc receptor common γ chain deficient (FcRγ(-/-) ) and wild-type mice were immunized with a tandem repeat of M2e presented on virus-like particles (M2e5x VLP). Levels of M2e-specific antibodies that were induced in FcRγ(-/-) mice after immunization with M2e5x VLP were similar to those in wild-type mice. In addition, M2e antibodies induced in FcRγ(-/-) mice were found to be equally protective as those induced in wild-type mice. However, M2e5x VLP-immunized FcRγ(-/-) mice were not well protected, as shown by severe weight loss, higher lung viral titres and interleukin-6 inflammatory cytokine production upon influenza virus challenge compared with M2e5x VLP-immunized wild-type mice. Importantly, FcRγ(-/-) mice that were immunized with inactivated influenza virus induced haemagglutination inhibition activity and were well protected without a significant weight loss. Interestingly, interferon-γ-producing CD4 T and CD8 T cells were found to be prevalent in lungs from M2e5x VLP-immunized FcRγ(-/-) mice, which appeared to be correlated with a faster recovery after infection. These results indicate that Fc receptors play a primary role in conferring M2e-specific antibody-mediated protection whereas T cells may contribute to the recovery at later stages of infection.
Collapse
Affiliation(s)
- Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Wang W, Huang B, Jiang T, Wang X, Qi X, Tan W, Ruan L. Maximal immune response and cross protection by influenza virus nucleoprotein derived from E. coli using an optimized formulation. Virology 2014; 468-470:265-273. [PMID: 25213406 DOI: 10.1016/j.virol.2014.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/11/2014] [Accepted: 08/09/2014] [Indexed: 12/16/2022]
Abstract
The highly conserved internal nucleoprotein (NP) is a promising antigen to develop a universal influenza A virus vaccine. In this study, mice were injected intramuscularly with Escherichia coli-derived NP protein alone or in combination with adjuvant alum (Al(OH)3), CpG or both. The results showed that the NP protein formulated with adjuvant was effective in inducing a protective immune response. Additionally, the adjuvant efficacy of Al(OH)3 was stronger than that of CpG. Optimal immune responses were observed in BALB/c mice immunized with a combination of NP protein plus Al(OH)3 and CpG. These mice also showed maximal resistance following challenge with influenza A virus PR8 strain. Most importantly, 10 µg NP formulated with Al(OH)3 and CpG induced higher protection than did 90 µg NP. These findings indicated that a combination of Al(OH)3 and CpG may be an efficient adjuvant in the NP formulation.
Collapse
Affiliation(s)
- Wenling Wang
- National Institute for Viral Disease Control & Prevention, Chinese Center for Disease Control and Prevention (China CDC), 155# Chang Bai Road, Chang Ping District, Beijing 102206, China
| | - Baoying Huang
- National Institute for Viral Disease Control & Prevention, Chinese Center for Disease Control and Prevention (China CDC), 155# Chang Bai Road, Chang Ping District, Beijing 102206, China
| | - Tao Jiang
- National Institute for Viral Disease Control & Prevention, Chinese Center for Disease Control and Prevention (China CDC), 155# Chang Bai Road, Chang Ping District, Beijing 102206, China
| | - Xiuping Wang
- National Institute for Viral Disease Control & Prevention, Chinese Center for Disease Control and Prevention (China CDC), 155# Chang Bai Road, Chang Ping District, Beijing 102206, China
| | - Xiangrong Qi
- National Institute for Viral Disease Control & Prevention, Chinese Center for Disease Control and Prevention (China CDC), 155# Chang Bai Road, Chang Ping District, Beijing 102206, China
| | - Wenjie Tan
- National Institute for Viral Disease Control & Prevention, Chinese Center for Disease Control and Prevention (China CDC), 155# Chang Bai Road, Chang Ping District, Beijing 102206, China
| | - Li Ruan
- National Institute for Viral Disease Control & Prevention, Chinese Center for Disease Control and Prevention (China CDC), 155# Chang Bai Road, Chang Ping District, Beijing 102206, China.
| |
Collapse
|
41
|
van de Sandt CE, Kreijtz JHCM, Geelhoed-Mieras MM, Vogelzang-van Trierum SE, Nieuwkoop NJ, van de Vijver DAMC, Fouchier RAM, Osterhaus ADME, Morein B, Rimmelzwaan GF. Novel G3/DT adjuvant promotes the induction of protective T cells responses after vaccination with a seasonal trivalent inactivated split-virion influenza vaccine. Vaccine 2014; 32:5614-23. [PMID: 25140929 DOI: 10.1016/j.vaccine.2014.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/17/2014] [Accepted: 08/06/2014] [Indexed: 12/20/2022]
Abstract
Vaccines used against seasonal influenza are poorly effective against influenza A viruses of novel subtypes that may have pandemic potential. Furthermore, pre(pandemic) influenza vaccines are poorly immunogenic, which can be overcome by the use of adjuvants. A limited number of adjuvants has been approved for use in humans, however there is a need for alternative safe and effective adjuvants that can enhance the immunogenicity of influenza vaccines and that promote the induction of broad-protective T cell responses. Here we evaluated a novel nanoparticle, G3, as an adjuvant for a seasonal trivalent inactivated influenza vaccine in a mouse model. The G3 adjuvant was formulated with or without steviol glycosides (DT, for diterpenoid). The use of both formulations enhanced the virus-specific antibody response to all three vaccine strains considerably. The adjuvants were well tolerated without any signs of discomfort. To assess the protective potential of the vaccine-induced immune responses, an antigenically distinct influenza virus strain, A/Puerto Rico/8/34 (A/PR/8/34), was used for challenge infection. The vaccine-induced antibodies did not cross-react with strain A/PR/8/34 in HI and VN assays. However, mice immunized with the G3/DT-adjuvanted vaccine were partially protected against A/PR/8/34 infection, which correlated with the induction of anamnestic virus-specific CD8(+) T cell responses that were not observed with the use of G3 without DT. Both formulations induced maturation of human dendritic cells and promoted antigen presentation to a similar extent. In conclusion, G3/DT is a promising adjuvant formulation that not only potentiates the antibody response induced by influenza vaccines, but also induces T cell immunity which could afford broader protection against antigenically distinct influenza viruses.
Collapse
Affiliation(s)
| | - Joost H C M Kreijtz
- Department of Viroscience, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | | - Nella J Nieuwkoop
- Department of Viroscience, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | - Ron A M Fouchier
- Department of Viroscience, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Albert D M E Osterhaus
- Department of Viroscience, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands; ViroClinics Biosciences BV, Marconistraat 16, 3029 AK Rotterdam, The Netherlands
| | - Bror Morein
- Infectious Diseases Department of Medical Sciences, Uppsala University, MoreinX, Dag Hammarskjöldsväg 34 A, 751 83 Uppsala, Sweden
| | - Guus F Rimmelzwaan
- Department of Viroscience, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands; ViroClinics Biosciences BV, Marconistraat 16, 3029 AK Rotterdam, The Netherlands.
| |
Collapse
|
42
|
Noisumdaeng P, Pooruk P, Prasertsopon J, Assanasen S, Kitphati R, Auewarakul P, Puthavathana P. Homosubtypic and heterosubtypic antibodies against highly pathogenic avian influenza H5N1 recombinant proteins in H5N1 survivors and non-H5N1 subjects. Virology 2014; 454-455:254-62. [PMID: 24725952 DOI: 10.1016/j.virol.2014.02.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/03/2014] [Accepted: 02/22/2014] [Indexed: 01/15/2023]
Abstract
Six recombinant vaccinia viruses containing HA, NA, NP, M or NS gene insert derived from a highly pathogenic avian influenza H5N1 virus, and the recombinant vaccinia virus harboring plasmid backbone as the virus control were constructed. The recombinant proteins were characterized for their expression and subcellular locations in TK(-) cells. Antibodies to the five recombinant proteins were detected in all 13 sequential serum samples collected from four H5N1 survivors during four years of follow-up; and those directed to rVac-H5 HA and rVac-NA proteins were found in higher titers than those directed to the internal proteins as revealed by indirect immunofluorescence assay. Although all 28 non-H5N1 subjects had no neutralizing antibodies against H5N1 virus, they did have cross-reactive antibodies to those five recombinant proteins. A significant increase in cross-reactive antibody titer to rVac-H5 HA and rVac-NA was found in paired blood samples from patients infected with the 2009 pandemic virus.
Collapse
Affiliation(s)
- Pirom Noisumdaeng
- Siriraj Influenza Cooperative Research Center, Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok 10700, Thailand; Center for Emerging and Neglected Infectious Disease, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Phisanu Pooruk
- Siriraj Influenza Cooperative Research Center, Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok 10700, Thailand
| | - Jarunee Prasertsopon
- Siriraj Influenza Cooperative Research Center, Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok 10700, Thailand
| | - Susan Assanasen
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok 10700, Thailand
| | - Rungrueng Kitphati
- Department of Disease Control, Ministry of Public Health, Nonthaburi 11000, Thailand
| | - Prasert Auewarakul
- Siriraj Influenza Cooperative Research Center, Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok 10700, Thailand; Center for Emerging and Neglected Infectious Disease, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Pilaipan Puthavathana
- Siriraj Influenza Cooperative Research Center, Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok 10700, Thailand; Center for Emerging and Neglected Infectious Disease, Mahidol University, Nakhon Pathom 73170, Thailand.
| |
Collapse
|
43
|
van de Sandt CE, Kreijtz JHCM, de Mutsert G, Geelhoed-Mieras MM, Hillaire MLB, Vogelzang-van Trierum SE, Osterhaus ADME, Fouchier RAM, Rimmelzwaan GF. Human cytotoxic T lymphocytes directed to seasonal influenza A viruses cross-react with the newly emerging H7N9 virus. J Virol 2014; 88:1684-93. [PMID: 24257602 PMCID: PMC3911609 DOI: 10.1128/jvi.02843-13] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/12/2013] [Indexed: 01/05/2023] Open
Abstract
In February 2013, zoonotic transmission of a novel influenza A virus of the H7N9 subtype was reported in China. Although at present no sustained human-to-human transmission has been reported, a pandemic outbreak of this H7N9 virus is feared. Since neutralizing antibodies to the hemagglutinin (HA) globular head domain of the virus are virtually absent in the human population, there is interest in identifying other correlates of protection, such as cross-reactive CD8(+) T cells (cytotoxic T lymphocytes [CTLs]) elicited during seasonal influenza A virus infections. These virus-specific CD8(+) T cells are known to recognize conserved internal proteins of influenza A viruses predominantly, but it is unknown to what extent they cross-react with the newly emerging H7N9 virus. Here, we assessed the cross-reactivity of seasonal H3N2 and H1N1 and pandemic H1N1 influenza A virus-specific polyclonal CD8(+) T cells, obtained from HLA-typed study subjects, with the novel H7N9 virus. The cross-reactivity of CD8(+) T cells to H7N9 variants of known influenza A virus epitopes and H7N9 virus-infected cells was determined by their gamma interferon (IFN-γ) response and lytic activity. It was concluded that, apart from recognition of individual H7N9 variant epitopes, CD8(+) T cells to seasonal influenza viruses display considerable cross-reactivity with the novel H7N9 virus. The presence of these cross-reactive CD8(+) T cells may afford some protection against infection with the new virus.
Collapse
MESH Headings
- Adult
- Amino Acid Sequence
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Cells, Cultured
- China/epidemiology
- Cross Protection
- Cross Reactions
- Disease Outbreaks
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Humans
- Influenza A Virus, H1N1 Subtype/chemistry
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/chemistry
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/chemistry
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/isolation & purification
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Influenza, Human/virology
- Interferon-gamma/immunology
- Male
- Middle Aged
- Molecular Sequence Data
- Seasons
- Sequence Alignment
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/virology
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ron A. M. Fouchier
- Viroscience Laboratory, Erasmus MC, Rotterdam, The Netherlands
- ViroClinics Biosciences BV, Rotterdam, The Netherlands
| | - Guus F. Rimmelzwaan
- Viroscience Laboratory, Erasmus MC, Rotterdam, The Netherlands
- ViroClinics Biosciences BV, Rotterdam, The Netherlands
| |
Collapse
|
44
|
Epstein SL. Control of influenza virus infection by immunity to conserved viral features. Expert Rev Anti Infect Ther 2014; 1:627-38. [PMID: 15482160 DOI: 10.1586/14787210.1.4.627] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Influenza has circulated among humans for centuries and kills more people than many newly emerging diseases. The present methods for control of influenza are not adequate, especially for dealing with a pandemic. In the face of a rapidly spreading outbreak, a race to isolate the virus and prepare a vaccine would probably not succeed in time to avoid great losses. Thus, additional anti-infection strategies are needed. Broad cross-protection against widely divergent influenza A subtypes is readily achieved in animals by several means of immunization. How does cross-protection work in animals, and can we apply what we have learned about it to induce broad cross-protection in humans?
Collapse
Affiliation(s)
- Suzanne L Epstein
- Laboratory of Immunology and Developmental Biology, Division of Cellular and Gene Therapies, HFM-730, Office of Cellular, Tissue and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, USA.
| |
Collapse
|
45
|
|
46
|
Abstract
Neuraminidase (NA) is the second most abundant influenza surface glycoprotein and contributes to virus replication in several ways, most notably by removing sialic acids from the host and viral glycoproteins, releasing newly formed virus particles from infected cells. Antibodies that block this enzyme activity restrict virus replication in vitro. This chapter describes foundational epidemiologic and human influenza challenge studies that provide evidence of an association between NA inhibiting antibodies and resistance to disease. Mouse challenge studies show that while NA immunity is infection-permissive, NA-specific antibodies attenuate infection and prevent severe disease. NA immunity is most effective against homologous viruses but there is substantial protection against viruses with a heterologous NA (different lineage within a NA subtype). Monoclonal antibodies specific for conserved antigenic domains of subtype N1 protect against seasonal and pandemic H1N1 as well as H5N1 virus challenge. Clinical studies demonstrate that licensed seasonal vaccines contain immunogenic amounts of NA, but the contribution of this immunity to vaccine efficacy is currently not known. New types of influenza vaccines could be designed to elicit NA immunity. Because NA induces heterologous immunity, it could be an important constituent of universal influenza vaccines that aim to protect against unexpected emerging viruses.
Collapse
Affiliation(s)
- Maryna C Eichelberger
- Division of Viral Products, Office of Vaccine Research and Review, Center for Biologics Research and Regulation, US Food and Drug Administration, HFM445, Silver Spring, MD, 20892, USA,
| | | |
Collapse
|
47
|
Salk HM, Haralambieva IH, Ovsyannikova IG, Goergen KM, Poland GA. Granzyme B ELISPOT assay to measure influenza-specific cellular immunity. J Immunol Methods 2013; 398-399:44-50. [PMID: 24055591 DOI: 10.1016/j.jim.2013.09.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 08/29/2013] [Accepted: 09/04/2013] [Indexed: 12/13/2022]
Abstract
The immunogenicity and efficacy of influenza vaccination are markedly lower in the elderly. Granzyme B (GrzB), quantified in fresh cell lysates, has been suggested to be a marker of cytotoxic T lymphocyte (CTL) response and a predictor of influenza illness among vaccinated older individuals. We have developed an influenza-specific GrzB ELISPOT assay using cryopreserved PBMCs. This method was tested on 106 healthy older subjects (ages 50-74) at baseline (Day 0) and three additional time points post-vaccination (Day 3, Day 28, Day 75) with influenza A/H1N1-containing vaccine. No significant difference was seen in GrzB response between any of the time points, although influenza-specific GrzB response appears to be elevated at all post-vaccination time points. There was no correlation between GrzB response and hemagglutination inhibition (HAI) titers, indicating no relationship between the cytolytic activity and humoral antibody levels in this cohort. Additionally, a significant negative correlation between GrzB response and age was observed. These results reveal a reduction in influenza-specific GrzB response as one ages. In conclusion, we have developed and optimized an influenza-specific ELISPOT assay for use with frozen cells to quantify the CTL-specific serine protease GrzB, as a measure of cellular immunity after influenza vaccination.
Collapse
|
48
|
Swinkels WJC, Hoeboer J, Sikkema R, Vervelde L, Koets ADP. Vaccination induced antibodies to recombinant avian influenza A virus M2 protein or synthetic M2e peptide do not bind to the M2 protein on the virus or virus infected cells. Virol J 2013; 10:206. [PMID: 23800100 PMCID: PMC3701469 DOI: 10.1186/1743-422x-10-206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 06/17/2013] [Indexed: 11/11/2022] Open
Abstract
Background Influenza viruses are characterized by their highly variable surface proteins HA and NA. The third surface protein M2 is a nearly invariant protein in all Influenza A strains. Despite extensive studies in other animal models, this study is the first to describe the use of recombinant M2 protein and a peptide coding for the extracellular part of the M2 protein (M2e) to vaccinate poultry. Methods Four groups of layer chickens received a prime-boost vaccination with recombinant M2 protein, M2e, a tetrameric construct from M2e peptide bound to streptavidin and a control tetrameric construct formulated with Stimune adjuvant. Results We determined the M2-specific antibody (Ab) responses in the serum before vaccination, three weeks after vaccination and two weeks after booster, at days 21, 42 and 56 of age. The group vaccinated with the M2 protein in combination with Stimune adjuvant showed a significant Ab response to the complete M2 protein as compared to the other groups. In addition an increased Ab response to M2e peptide was found in the group vaccinated with the M2e tetrameric construct. None of the vaccinated animals showed seroconversion to AI in a commercial ELISA. Finally no Ab’s were found that bound to M2 expressed on in vitro AI infected MDCK cells. Conclusion Although Ab’s are formed against the M2 protein and to Streptavidin bound M2e peptide in a tetrameric conformation these Ab’s do not recognize of M2 on the virus or on infected cells.
Collapse
Affiliation(s)
- Willem J C Swinkels
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 Utrecht CL, The Netherlands
| | | | | | | | | |
Collapse
|
49
|
Abstract
Vaccination is the primary strategy for prevention and control of influenza. The surface hemagglutinin (HA) protein of the influenza virus contains two structural elements (head and stalk) that differ in their potential utility as vaccine targets. The head of the HA protein is the primary target of antibodies that confer protective immunity to influenza viruses. The underlying health status, age, and gene polymorphisms of vaccine recipients and, just as importantly, the extent of the antigenic match between the viruses in the vaccine and those that are circulating modulate influenza vaccine protection. Vaccine adjuvants and live attenuated influenza vaccine improve the breadth of immunity to seasonal and pandemic virus strains. Eliciting antibodies against the conserved HA stem region that cross-react with HAs within influenza virus types or subtypes would allow for the development of a universal influenza vaccine. The highly complex network of interactions generated after influenza infection and vaccination can be studied with the use of systems biology tools, such as DNA microarray chips. The use of systems vaccinology has allowed for the generation of gene expression signatures that represent key transcriptional differences between asymptomatic and symptomatic host responses to influenza infection. Additionally, the use of systems vaccinology tools have resulted in the identification of novel surrogate gene markers that are predictors of the magnitude of host responses to vaccines, which is critical to both vaccine development and public health. Identifying associations between variations in vaccine immune responses and gene polymorphisms is critical in the development of universal influenza vaccines.
Collapse
Affiliation(s)
| | - Matthew J Fenton
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
50
|
Subbarao K, Matsuoka Y. The prospects and challenges of universal vaccines for influenza. Trends Microbiol 2013; 21:350-8. [PMID: 23685068 DOI: 10.1016/j.tim.2013.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/04/2013] [Accepted: 04/15/2013] [Indexed: 12/25/2022]
Abstract
Vaccination is the most effective way to reduce the impact of epidemic as well as pandemic influenza. However, the licensed inactivated influenza vaccine induces strain-specific immunity and must be updated annually. When novel viruses appear, matched vaccines are not likely to be available in time for the first wave of a pandemic. Yet, the enormous diversity of influenza A viruses in nature makes it impossible to predict which subtype or strain will cause the next pandemic. Several recent scientific advances have generated renewed enthusiasm and hope for universal vaccines that will induce broad protection from a range of influenza viruses.
Collapse
Affiliation(s)
- Kanta Subbarao
- National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA.
| | | |
Collapse
|