1
|
Sanchez-Martinez ZV, Alpuche-Lazcano SP, Stuible M, Akache B, Renner TM, Deschatelets L, Dudani R, Harrison BA, McCluskie MJ, Hrapovic S, Blouin J, Wang X, Schuller M, Cui K, Cho JY, Durocher Y. SARS-CoV-2 spike-based virus-like particles incorporate influenza H1/N1 antigens and induce dual immunity in mice. Vaccine 2024; 42:126463. [PMID: 39481241 DOI: 10.1016/j.vaccine.2024.126463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 11/02/2024]
Abstract
A vaccine effective against both SARS-CoV-2 and influenza A (IAV) viruses could represent a cost-effective strategy to reduce their combined public health burden as well as potential complications arising from co-infection. Based on previous findings that full-length SARS-CoV-2 spike (S) expression can induce high-level, enveloped VLP (eVLP) production in CHO cells, we tested whether IAV H1N1 hemagglutinin (H1) and neuraminidase (N1) could also be displayed on these particles. We found that co-incorporation of the IAV surface antigens in spike VLPs (S-VLPs) was highly efficient: upon transient co-expression of S + H1 or S + H1 + N1 in CHO cells, the resulting VLPs contained similar amounts of the SARS-CoV-2 S and IAV antigens. The self-assembled bivalent (S/H1) and trivalent (S/H1/N1) VLPs released into the culture media were purified by single-step chromatography using a S-VLP affinity resin. Western blot analysis and immuno‑gold labeling transmission electron microscopy (TEM) of purified VLPs confirmed the coexistence of S, H1 and N1 antigens in the same particles. Finally, we demonstrated that two doses of adjuvanted bivalent and trivalent VLPs elicit specific functional antibodies and cellular immunity in a mouse model, suggesting potential for combined SARS-CoV-2/IAV vaccine development.
Collapse
Affiliation(s)
- Zalma V Sanchez-Martinez
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada.; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada.; Current address: Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, Cuernavaca Morelos (CP 62250), Mexico
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Bassel Akache
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Tyler M Renner
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Lise Deschatelets
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Renu Dudani
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Blair A Harrison
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada
| | - Sabahudin Hrapovic
- Aquatic and Crop Resources Development Research Centre, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Julie Blouin
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Xinyu Wang
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Matthew Schuller
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Kai Cui
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Jae-Young Cho
- Quantum and Nanotechnologies Research Centre, National Research Council Canada, 11421 Saskatchewan Drive, Edmonton, AB T6G 2M9, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada.; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada..
| |
Collapse
|
2
|
Hoxie I, Vasilev K, Clark JJ, Bushfield K, Francis B, Loganathan M, Campbell JD, Yu D, Guan L, Gu C, Fan S, Tompkins SM, Neumann G, Kawaoka Y, Krammer F. A recombinant N2 neuraminidase-based CpG 1018® adjuvanted vaccine provides protection against challenge with heterologous influenza viruses in mice and hamsters. Vaccine 2024; 42:126269. [PMID: 39241354 DOI: 10.1016/j.vaccine.2024.126269] [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: 07/09/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
Recombinant influenza virus neuraminidase (NA) is a promising broadly protective influenza vaccine candidate. However, the recombinant protein alone is not sufficient to induce durable and protective immune responses and requires the coadministration of immunostimulatory molecules. Here, we evaluated the immunogenicity and cross-protective potential of a recombinant influenza virus N2 neuraminidase vaccine construct, adjuvanted with a toll-like receptor 9 (TLR9) agonist (CpG 1018® adjuvant), and alum. The combination of CpG 1018 adjuvant and alum induced a balanced and robust humoral and T-cellular immune response against the NA, which provided protection and reduced morbidity against homologous and heterologous viral challenges in mouse and hamster models. This study supports Syrian hamsters as a useful complementary animal model to mice for pre-clinical evaluation of influenza virus vaccines.
Collapse
Affiliation(s)
- Irene Hoxie
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirill Vasilev
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordan J Clark
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaitlyn Bushfield
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Francis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Dong Yu
- Dynavax Technologies Corporation, Emeryville, CA, USA
| | - Lizheng Guan
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Chunyang Gu
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Shufang Fan
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - S Mark Tompkins
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; Center for Influenza Disease and Emergence Response (CIDER), University of Georgia, Athens, GA 30602, USA; Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Gabriele Neumann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA; Department of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan; The University of Tokyo Pandemic Preparedness, Infection and Advanced research center (UTOPIA), Tokyo, Japan
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Guzman Ruiz L, Zollner AM, Hoxie I, Küchler J, Hausjell C, Mesurado T, Krammer F, Jungbauer A, Pereira Aguilar P, Klausberger M, Grabherr R. Enhancing NA immunogenicity through novel VLP designs. Vaccine 2024; 42:126270. [PMID: 39197219 DOI: 10.1016/j.vaccine.2024.126270] [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: 06/27/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Current influenza virus vaccines poorly display key neuraminidase (NA) epitopes and do not robustly induce NA-reactive antibodies; instead, they focus on the induction of hemagglutinin (HA)-reactive antibodies. Next-generation influenza vaccines should be optimized in order to activate NA-reactive B cells and to induce a broadly cross-reactive and protective antibody response. We aimed at enhancing the immunogenicity of the NA on vaccines by two strategies: (i) modifying the HA:NA ratio of the vaccine preparation and (ii) exposing epitopes on the lateral surface or beneath the head of the NA by extending the NA stalk. The H1N1 glycoproteins from the influenza virus A/California/04/2009 strain were displayed on human immunodeficiency virus 1 (HIV-1) gag-based virus-like particles (VLP). Using the baculovirus insect cell expression system, we biased the quantity of surface glycoproteins employing two different promoters, the very late baculovirus p10 promoter and the early and late gp64 promoter. This led to a 1:1 to 2:1 HA:NA ratio, which was approximately double or triple the amount of NA as present on the wild-type influenza A virus (HA:NA ratio 3:1 to 5:1). Furthermore, by insertion of 15 amino acids from the A-New York/61/2012 strain (NY12) which prolongates the NA stalk (NA long stalk; NA-LS), we intended to improve the accessibility of the NA. Six different types of VLPs were produced and purified using a platform downstream process based on Capto-Core 700™ followed by Capto-Heparin™ affinity chromatography combined with ultracentrifugation. These VLPs were then tested in a mouse model. Robust titers of antibodies that inhibit the neuraminidase activity were elicited even after vaccination with two low doses (0.3 μg) of the H1N1 VLPs without compromising the anti-HA responses. In conclusion, our results demonstrate the feasibility of the two developed strategies to retain HA immunogenicity and improve NA immunogenicity as a future influenza vaccine candidate.
Collapse
Affiliation(s)
- Leticia Guzman Ruiz
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria; University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Alexander M Zollner
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Irene Hoxie
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, Gustave L. Levy Place, 10029-5674 New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jan Küchler
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Christina Hausjell
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria
| | - Tomas Mesurado
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, Gustave L. Levy Place, 10029-5674 New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Alois Jungbauer
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria
| | - Patricia Pereira Aguilar
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria
| | - Miriam Klausberger
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria
| | - Reingard Grabherr
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
4
|
Septer KM, Heinly TA, Sim DG, Patel DR, Roder AE, Wang W, Chung M, Johnson KEE, Ghedin E, Sutton TC. Vaccine-induced NA immunity decreases viral shedding, but does not disrupt chains of airborne transmission for the 2009 pandemic H1N1 virus in ferrets. mBio 2024; 15:e0216124. [PMID: 39248566 PMCID: PMC11481891 DOI: 10.1128/mbio.02161-24] [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: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024] Open
Abstract
Split-virion-inactivated influenza vaccines are formulated based on viral hemagglutinin content. These vaccines also contain the viral neuraminidase (NA) protein, but NA content is not standardized and varies between manufacturers. In clinical studies and animal models, antibodies directed toward NA reduced disease severity and viral load; however, the impact of vaccine-induced NA immunity on airborne transmission of influenza A viruses is not well characterized. Therefore, we evaluated if vaccination against NA could disrupt chains of airborne transmission for the 2009 pandemic H1N1 virus in ferrets. Immunologically naïve donor ferrets were infected with the 2009 pandemic H1N1 virus and then paired in transmission cages with mock- or NA-vaccinated respiratory contacts. The mock- and NA-vaccinated animals were then monitored daily for infection, and once infected, these animals were paired with a naive secondary respiratory contact. In these studies, all mock- and NA-vaccinated animals became infected; however, NA-vaccinated animals shed significantly less virus for fewer days relative to mock-vaccinated animals. For the secondary contacts, 6/6 and 5/6 animals became infected after exposure to mock- and NA-vaccinated animals, respectively. To determine if vaccine-induced immune pressure selected for escape variants, we sequenced viruses recovered from ferrets. No mutations in NA became enriched during transmission. These findings indicate that despite reducing viral load, vaccine-induced NA immunity does not prevent infection during continuous airborne exposure and subsequent onward airborne transmission of the 2009 pandemic H1N1 virus. IMPORTANCE In humans and animal models, immunity against neuraminidase (NA) reduces disease severity and viral replication during influenza infection. However, we have a limited understanding of the impact of NA immunity on viral transmission. Using chains of airborne transmission in ferrets as a strategy to simulate a more natural route of infection, we assessed if vaccine-induced NA immunity could disrupt transmission of the 2009 pandemic H1N1 virus. The 2009 pandemic H1N1 virus transmitted efficiently through chains of transmission in the presence of NA immunity, but NA-vaccinated animals shed significantly less virus and had accelerated viral clearance. To determine if immune pressure led to the generation of escape variants, viruses in ferret nasal wash samples were sequenced, and no mutations in NA were identified. These findings demonstrate that vaccine-induced NA immunity is not sufficient to prevent infection via airborne exposure and onward airborne transmission of the 2009 pandemic H1N1 virus.
Collapse
Affiliation(s)
- K. M. Septer
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Emory-UGA Center of Excellence of Influenza Research and Response (CEIRR), University Park, Pennsylvania, USA
| | - T. A. Heinly
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Emory-UGA Center of Excellence of Influenza Research and Response (CEIRR), University Park, Pennsylvania, USA
| | - D. G. Sim
- Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania, USA
| | - D. R. Patel
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Emory-UGA Center of Excellence of Influenza Research and Response (CEIRR), University Park, Pennsylvania, USA
| | - A. E. Roder
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - W. Wang
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - M. Chung
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - K. E. E. Johnson
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - E. Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - T. C. Sutton
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
- Emory-UGA Center of Excellence of Influenza Research and Response (CEIRR), University Park, Pennsylvania, USA
| |
Collapse
|
5
|
Wang M, Gao Y, Shen C, Yang W, Peng Q, Cheng J, Shen HM, Yang Y, Gao GF, Shi Y. A human monoclonal antibody targeting the monomeric N6 neuraminidase confers protection against avian H5N6 influenza virus infection. Nat Commun 2024; 15:8871. [PMID: 39402031 PMCID: PMC11473554 DOI: 10.1038/s41467-024-53301-6] [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: 05/17/2024] [Accepted: 10/09/2024] [Indexed: 10/17/2024] Open
Abstract
The influenza neuraminidase (NA) is a potential target for the development of a next-generation influenza vaccine, but its antigenicity is not well understood. Here, we isolate an anti-N6 human monoclonal antibody, named 18_14D, from an H5N6 avian influenza virus (AIV) infected patient. The antibody weakly inhibits enzymatic activity but confers protection in female mice, mainly via ADCC function. The cryo-EM structure shows that 18_14D binds to a unique epitope on the lateral surface of the N6 tetramer, preventing the formation of tightly closed NA tetramers. These findings contribute to the molecular understanding of protective immune responses to NA of AIVs in humans and open an avenue for the rational design of NA-based vaccines.
Collapse
Affiliation(s)
- Min Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen, China
| | - Yuan Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Chenguang Shen
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen, China
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Southern Medical University, Guangzhou, China
| | - Wei Yang
- Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qi Peng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Jinlong Cheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen, China.
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
- Medical School, University of Chinese Academy of Sciences, Beijing, China.
- Beijing Life Science Academy, Beijing, China.
| | - Yi Shi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital, Shenzhen, China.
- Medical School, University of Chinese Academy of Sciences, Beijing, China.
- Beijing Life Science Academy, Beijing, China.
- Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
6
|
Vatzia E, Paudyal B, Dema B, Carr BV, Sedaghat-Rostami E, Gubbins S, Sharma B, Moorhouse E, Morris S, Ulaszewska M, MacLoughlin R, Salguero FJ, Gilbert SC, Tchilian E. Aerosol immunization with influenza matrix, nucleoprotein, or both prevents lung disease in pig. NPJ Vaccines 2024; 9:188. [PMID: 39397062 PMCID: PMC11471855 DOI: 10.1038/s41541-024-00989-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
Current influenza vaccines are strain-specific and require frequent updates to combat new strains, making a broadly protective influenza vaccine (BPIV) highly desirable. A promising strategy is to induce T-cell responses against internal proteins conserved across influenza strains. In this study, pH1N1 pre-exposed pigs were immunized by aerosol using viral vectored vaccines (ChAdOx2 and MVA) expressing matrix (M1) and nucleoprotein (NP). Following H3N2 challenge, all immunizations (M1, NP or NPM1) reduced lung pathology, but M1 alone offered the greatest protection. NP or NPM1 immunization induced both T-cell and antibody responses. M1 immunization generated no detectable antibodies but elicited M1-specific T-cell responses, suggesting T cell-mediated protection. Additionally, a single aerosol immunization with the ChAdOx vaccine encoding M1, NP and neuraminidase reduced lung pathology. These findings provide insights into BPIV development using a relevant large natural host, the pig.
Collapse
Affiliation(s)
| | | | - Barbara Dema
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | | | | | | - Susan Morris
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marta Ulaszewska
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | - Sarah C Gilbert
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | |
Collapse
|
7
|
Leonard RA, Burke KN, Spreng RL, Macintyre AN, Tam Y, Alameh MG, Weissman D, Heaton NS. Improved influenza vaccine responses after expression of multiple viral glycoproteins from a single mRNA. Nat Commun 2024; 15:8712. [PMID: 39379405 PMCID: PMC11461824 DOI: 10.1038/s41467-024-52940-z] [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: 05/24/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
Influenza viruses cause substantial morbidity and mortality every year despite seasonal vaccination. mRNA-based vaccines have the potential to elicit more protective immune responses, but for maximal breadth and durability, it is desirable to deliver both the viral hemagglutinin and neuraminidase glycoproteins. Delivering multiple antigens individually, however, complicates manufacturing and increases cost, thus it would be beneficial to express both proteins from a single mRNA. Here, we develop an mRNA genetic configuration that allows the simultaneous expression of unmodified, full-length NA and HA proteins from a single open reading frame. We apply this approach to glycoproteins from contemporary influenza A and B viruses and, after vaccination, observe high levels of functional antibodies and protection from disease in female mouse and male ferret challenge models. This approach may further efforts to utilize mRNA technology to improve seasonal vaccine efficacy by efficiently delivering multiple viral antigens simultaneously and in their native state.
Collapse
MESH Headings
- Animals
- Ferrets
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Female
- Mice
- Male
- Neuraminidase/immunology
- Neuraminidase/genetics
- Antibodies, Viral/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Humans
- Mice, Inbred BALB C
- Influenza B virus/immunology
- Influenza B virus/genetics
- Influenza A virus/immunology
- Influenza A virus/genetics
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Influenza, Human/virology
- Glycoproteins/immunology
- Glycoproteins/genetics
- Viral Proteins/immunology
- Viral Proteins/genetics
- Antigens, Viral/immunology
- Antigens, Viral/genetics
- Vaccination/methods
Collapse
Affiliation(s)
- Rebecca A Leonard
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine Durham, Durham, NC, USA
| | - Kaitlyn N Burke
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine Durham, Durham, NC, USA
| | - Rachel L Spreng
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Andrew N Macintyre
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Ying Tam
- Acuitas Theraputics, Vancouver, BC, Canada
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Institute for RNA Innovation, Perelman School of Medicine, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, Institute for RNA Innovation, Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicholas S Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine Durham, Durham, NC, USA.
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
8
|
Perofsky AC, Huddleston J, Hansen CL, Barnes JR, Rowe T, Xu X, Kondor R, Wentworth DE, Lewis N, Whittaker L, Ermetal B, Harvey R, Galiano M, Daniels RS, McCauley JW, Fujisaki S, Nakamura K, Kishida N, Watanabe S, Hasegawa H, Sullivan SG, Barr IG, Subbarao K, Krammer F, Bedford T, Viboud C. Antigenic drift and subtype interference shape A(H3N2) epidemic dynamics in the United States. eLife 2024; 13:RP91849. [PMID: 39319780 PMCID: PMC11424097 DOI: 10.7554/elife.91849] [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] [Indexed: 09/26/2024] Open
Abstract
Influenza viruses continually evolve new antigenic variants, through mutations in epitopes of their major surface proteins, hemagglutinin (HA) and neuraminidase (NA). Antigenic drift potentiates the reinfection of previously infected individuals, but the contribution of this process to variability in annual epidemics is not well understood. Here, we link influenza A(H3N2) virus evolution to regional epidemic dynamics in the United States during 1997-2019. We integrate phenotypic measures of HA antigenic drift and sequence-based measures of HA and NA fitness to infer antigenic and genetic distances between viruses circulating in successive seasons. We estimate the magnitude, severity, timing, transmission rate, age-specific patterns, and subtype dominance of each regional outbreak and find that genetic distance based on broad sets of epitope sites is the strongest evolutionary predictor of A(H3N2) virus epidemiology. Increased HA and NA epitope distance between seasons correlates with larger, more intense epidemics, higher transmission, greater A(H3N2) subtype dominance, and a greater proportion of cases in adults relative to children, consistent with increased population susceptibility. Based on random forest models, A(H1N1) incidence impacts A(H3N2) epidemics to a greater extent than viral evolution, suggesting that subtype interference is a major driver of influenza A virus infection ynamics, presumably via heterosubtypic cross-immunity.
Collapse
MESH Headings
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- United States/epidemiology
- Influenza, Human/epidemiology
- Influenza, Human/virology
- Influenza, Human/immunology
- Humans
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Epidemics
- Antigenic Drift and Shift/genetics
- Child
- Adult
- Neuraminidase/genetics
- Neuraminidase/immunology
- Adolescent
- Child, Preschool
- Antigens, Viral/immunology
- Antigens, Viral/genetics
- Young Adult
- Evolution, Molecular
- Seasons
- Middle Aged
Collapse
Affiliation(s)
- Amanda C Perofsky
- Fogarty International Center, National Institutes of Health, Bethesda, United States
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, United States
| | - John Huddleston
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, United States
| | - Chelsea L Hansen
- Fogarty International Center, National Institutes of Health, Bethesda, United States
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, United States
| | - John R Barnes
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, United States
| | - Thomas Rowe
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, United States
| | - Xiyan Xu
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, United States
| | - Rebecca Kondor
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, United States
| | - David E Wentworth
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, United States
| | - Nicola Lewis
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Lynne Whittaker
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Burcu Ermetal
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Ruth Harvey
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Monica Galiano
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Rodney Stuart Daniels
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - John W McCauley
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Seiichiro Fujisaki
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kazuya Nakamura
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Noriko Kishida
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinji Watanabe
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hideki Hasegawa
- Influenza Virus Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sheena G Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Florian Krammer
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, United States
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Trevor Bedford
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, United States
- Department of Genome Sciences, University of Washington, Seattle, United States
- Howard Hughes Medical Institute, Seattle, United States
| | - Cécile Viboud
- Fogarty International Center, National Institutes of Health, Bethesda, United States
| |
Collapse
|
9
|
Pekarek MJ, Weaver EA. Influenza B Virus Vaccine Innovation through Computational Design. Pathogens 2024; 13:755. [PMID: 39338946 PMCID: PMC11434669 DOI: 10.3390/pathogens13090755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024] Open
Abstract
As respiratory pathogens, influenza B viruses (IBVs) cause a significant socioeconomic burden each year. Vaccine and antiviral development for influenza viruses has historically viewed IBVs as a secondary concern to influenza A viruses (IAVs) due to their lack of animal reservoirs compared to IAVs. However, prior to the global spread of SARS-CoV-2, the seasonal epidemics caused by IBVs were becoming less predictable and inducing more severe disease, especially in high-risk populations. Globally, researchers have begun to recognize the need for improved prevention strategies for IBVs as a primary concern. This review discusses what is known about IBV evolutionary patterns and the effect of the spread of SARS-CoV-2 on these patterns. We also analyze recent advancements in the development of novel vaccines tested against IBVs, highlighting the promise of computational vaccine design strategies when used to target both IBVs and IAVs and explain why these novel strategies can be employed to improve the effectiveness of IBV vaccines.
Collapse
Affiliation(s)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
10
|
Jones CH, Hauguel T, Beitelshees M, Davitt M, Welch V, Lindert K, Allen P, True JM, Dolsten M. Deciphering immune responses: a comparative analysis of influenza vaccination platforms. Drug Discov Today 2024; 29:104125. [PMID: 39097221 DOI: 10.1016/j.drudis.2024.104125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/21/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Influenza still poses a significant challenge due to its high mutation rates and the low effectiveness of traditional vaccines. At present, antibodies that neutralize the highly variable hemagglutinin antigen are a major driver of the observed variable protection. To decipher how influenza vaccines can be improved, an analysis of licensed vaccine platforms was conducted, contrasting the strengths and limitations of their different mechanisms of protection. Through this review, it is evident that these vaccines do not elicit the robust cellular immune response critical for protecting high-risk groups. Emerging platforms, such as RNA vaccines, that induce robust cellular responses that may be additive to the recognized mechanism of protection through hemagglutinin inhibition may overcome these constraints to provide broader, protective immunity. By combining both humoral and cellular responses, such platforms could help guide the future influenza vaccine development.
Collapse
Affiliation(s)
| | | | | | | | - Verna Welch
- Pfizer, Hudson Boulevard, New York, NY 10018, USA
| | | | - Pirada Allen
- Pfizer, Hudson Boulevard, New York, NY 10018, USA
| | - Jane M True
- Pfizer, Hudson Boulevard, New York, NY 10018, USA.
| | | |
Collapse
|
11
|
Luo J, Zhang M, Ye Q, Gao F, Xu W, Li B, Wang Q, Zhao L, Tan WS. A synthetic TLR4 agonist significantly increases humoral immune responses and the protective ability of an MDCK-cell-derived inactivated H7N9 vaccine in mice. Arch Virol 2024; 169:163. [PMID: 38990396 DOI: 10.1007/s00705-024-06082-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024]
Abstract
Antigenically divergent H7N9 viruses pose a potential threat to public health, with the poor immunogenicity of candidate H7N9 vaccines demonstrated in clinical trials underscoring the urgent need for more-effective H7N9 vaccines. In the present study, mice were immunized with various doses of a suspended-MDCK-cell-derived inactivated H7N9 vaccine, which was based on a low-pathogenic H7N9 virus, to assess cross-reactive immunity and cross-protection against antigenically divergent H7N9 viruses. We found that the CRX-527 adjuvant, a synthetic TLR4 agonist, significantly enhanced the humoral immune responses of the suspended-MDCK-cell-derived H7N9 vaccine, with significant antigen-sparing and immune-enhancing effects, including robust virus-specific IgG, hemagglutination-inhibiting (HI), neuraminidase-inhibiting (NI), and virus-neutralizing (VN) antibody responses, which are crucial for protection against influenza virus infection. Moreover, the CRX-527-adjuvanted H7N9 vaccine also elicited cross-protective immunity and cross-protection against a highly pathogenic H7N9 virus with a single vaccination. Notably, NI and VN antibodies might play an important role in cross-protection against lethal influenza virus infections. This study showed that a synthetic TLR4 agonist adjuvant has a potent immunopotentiating effect, which might be considered worth further development as a means of increasing vaccine effectiveness.
Collapse
Affiliation(s)
- Jian Luo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Institute of Biological Products, Shanghai, China
| | - Min Zhang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Qian Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Feixia Gao
- Shanghai Institute of Biological Products, Shanghai, China
| | - Wenting Xu
- Shanghai Institute of Biological Products, Shanghai, China
| | - Beibei Li
- Shanghai Institute of Biological Products, Shanghai, China
| | - Qi Wang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Liang Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
12
|
Kang H, Martinez MR, Aves KL, Okholm AK, Wan H, Chabot S, Malik T, Sander AF, Daniels R. Capsid virus-like particle display improves recombinant influenza neuraminidase antigen stability and immunogenicity in mice. iScience 2024; 27:110038. [PMID: 38883830 PMCID: PMC11179578 DOI: 10.1016/j.isci.2024.110038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 03/20/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Supplementing influenza vaccines with additional protective antigens such as neuraminidase (NA) is a promising strategy for increasing the breadth of the immune response. Here, we improved the immunogenicity and stability of secreted recombinant NA (rNA) tetramers by covalently conjugating them onto the surface of AP205 capsid virus-like particles (cVLPs) using a Tag/Catcher ligation system. cVLP display increased the induction of IgG2a subclass anti-NA antibodies, which exhibited cross-reactivity with an antigenically distant homologous NA. It also reduced the single dose rNA amounts needed for protection against viral challenge in mice, demonstrating a dose-sparing effect. Moreover, effective cVLP-display was achieved across different NA subtypes, even when the conjugation was performed shortly before administration. Notably, the rNA-cVLP immunogenicity was retained upon mixing or co-administering with commercial vaccines. These results highlight the potential of this approach for bolstering the protective immune responses elicited by influenza vaccines.
Collapse
Affiliation(s)
- Hyeog Kang
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Mira Rakic Martinez
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kara-Lee Aves
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Anna Kathrine Okholm
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Hongquan Wan
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Sylvie Chabot
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Tahir Malik
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Adam F Sander
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark
- AdaptVac, Ole Maaløes Vej 3, 2200 Copenhagen, Denmark
| | - Robert Daniels
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
13
|
Le Sage V, Rockey NC, French AJ, McBride R, McCarthy KR, Rigatti LH, Shephard MJ, Jones JE, Walter SG, Doyle JD, Xu L, Barbeau DJ, Wang S, Frizzell SA, Myerburg MM, Paulson JC, McElroy AK, Anderson TK, Vincent Baker AL, Lakdawala SS. Potential pandemic risk of circulating swine H1N2 influenza viruses. Nat Commun 2024; 15:5025. [PMID: 38871701 PMCID: PMC11176300 DOI: 10.1038/s41467-024-49117-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Influenza A viruses in swine have considerable genetic diversity and continue to pose a pandemic threat to humans due to a potential lack of population level immunity. Here we describe a pipeline to characterize and triage influenza viruses for their pandemic risk and examine the pandemic potential of two widespread swine origin viruses. Our analysis reveals that a panel of human sera collected from healthy adults in 2020 has no cross-reactive neutralizing antibodies against a α-H1 clade strain (α-swH1N2) but do against a γ-H1 clade strain. The α-swH1N2 virus replicates efficiently in human airway cultures and exhibits phenotypic signatures similar to the human H1N1 pandemic strain from 2009 (H1N1pdm09). Furthermore, α-swH1N2 is capable of efficient airborne transmission to both naïve ferrets and ferrets with prior seasonal influenza immunity. Ferrets with H1N1pdm09 pre-existing immunity show reduced α-swH1N2 viral shedding and less severe disease signs. Despite this, H1N1pdm09-immune ferrets that became infected via the air can still onward transmit α-swH1N2 with an efficiency of 50%. These results indicate that this α-swH1N2 strain has a higher pandemic potential, but a moderate level of impact since there is reduced replication fitness and pathology in animals with prior immunity.
Collapse
MESH Headings
- Animals
- Ferrets/virology
- Humans
- Swine
- Influenza, Human/virology
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Influenza, Human/blood
- Influenza, Human/transmission
- Orthomyxoviridae Infections/virology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/epidemiology
- Orthomyxoviridae Infections/transmission
- Orthomyxoviridae Infections/blood
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/isolation & purification
- Influenza A Virus, H1N2 Subtype/genetics
- Influenza A Virus, H1N2 Subtype/immunology
- Pandemics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Swine Diseases/virology
- Swine Diseases/epidemiology
- Swine Diseases/immunology
- Swine Diseases/transmission
- Swine Diseases/blood
- Female
- Virus Shedding
- Male
- Adult
- Virus Replication
Collapse
Affiliation(s)
- Valerie Le Sage
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nicole C Rockey
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Civil and Environmental Engineering, Duke University, Durham, NC, USA
| | - Andrea J French
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ryan McBride
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Kevin R McCarthy
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lora H Rigatti
- Division of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meredith J Shephard
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jennifer E Jones
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sydney G Walter
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joshua D Doyle
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lingqing Xu
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dominique J Barbeau
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shengyang Wang
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sheila A Frizzell
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael M Myerburg
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - James C Paulson
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Anita K McElroy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tavis K Anderson
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, Ames, IA, USA
| | - Amy L Vincent Baker
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, Ames, IA, USA
| | - Seema S Lakdawala
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Pascha MN, Ballegeer M, Roelofs MC, Meuris L, Albulescu IC, van Kuppeveld FJM, Hurdiss DL, Bosch BJ, Zeev-Ben-Mordehai T, Saelens X, de Haan CAM. Nanoparticle display of neuraminidase elicits enhanced antibody responses and protection against influenza A virus challenge. NPJ Vaccines 2024; 9:97. [PMID: 38821988 PMCID: PMC11143307 DOI: 10.1038/s41541-024-00891-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 05/20/2024] [Indexed: 06/02/2024] Open
Abstract
Current Influenza virus vaccines primarily induce antibody responses against variable epitopes in hemagglutinin (HA), necessitating frequent updates. However, antibodies against neuraminidase (NA) can also confer protection against influenza, making NA an attractive target for the development of novel vaccines. In this study, we aimed to enhance the immunogenicity of recombinant NA antigens by presenting them multivalently on a nanoparticle carrier. Soluble tetrameric NA antigens of the N1 and N2 subtypes, confirmed to be correctly folded by cryo-electron microscopy structural analysis, were conjugated to Mi3 self-assembling protein nanoparticles using the SpyTag-SpyCatcher system. Immunization of mice with NA-Mi3 nanoparticles induced higher titers of NA-binding and -inhibiting antibodies and improved protection against a lethal challenge compared to unconjugated NA. Additionally, we explored the co-presentation of N1 and N2 antigens on the same Mi3 particles to create a mosaic vaccine candidate. These mosaic nanoparticles elicited antibody titers that were similar or superior to the homotypic nanoparticles and effectively protected against H1N1 and H3N2 challenge viruses. The NA-Mi3 nanoparticles represent a promising vaccine candidate that could complement HA-directed approaches for enhanced potency and broadened protection against influenza A virus.
Collapse
Affiliation(s)
- M N Pascha
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - M Ballegeer
- VIB Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium
| | - M C Roelofs
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - L Meuris
- VIB Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium
| | - I C Albulescu
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - F J M van Kuppeveld
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - D L Hurdiss
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - B J Bosch
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - T Zeev-Ben-Mordehai
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - X Saelens
- VIB Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium.
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium.
| | - C A M de Haan
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
15
|
Perofsky AC, Huddleston J, Hansen C, Barnes JR, Rowe T, Xu X, Kondor R, Wentworth DE, Lewis N, Whittaker L, Ermetal B, Harvey R, Galiano M, Daniels RS, McCauley JW, Fujisaki S, Nakamura K, Kishida N, Watanabe S, Hasegawa H, Sullivan SG, Barr IG, Subbarao K, Krammer F, Bedford T, Viboud C. Antigenic drift and subtype interference shape A(H3N2) epidemic dynamics in the United States. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.02.23296453. [PMID: 37873362 PMCID: PMC10593063 DOI: 10.1101/2023.10.02.23296453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Influenza viruses continually evolve new antigenic variants, through mutations in epitopes of their major surface proteins, hemagglutinin (HA) and neuraminidase (NA). Antigenic drift potentiates the reinfection of previously infected individuals, but the contribution of this process to variability in annual epidemics is not well understood. Here we link influenza A(H3N2) virus evolution to regional epidemic dynamics in the United States during 1997-2019. We integrate phenotypic measures of HA antigenic drift and sequence-based measures of HA and NA fitness to infer antigenic and genetic distances between viruses circulating in successive seasons. We estimate the magnitude, severity, timing, transmission rate, age-specific patterns, and subtype dominance of each regional outbreak and find that genetic distance based on broad sets of epitope sites is the strongest evolutionary predictor of A(H3N2) virus epidemiology. Increased HA and NA epitope distance between seasons correlates with larger, more intense epidemics, higher transmission, greater A(H3N2) subtype dominance, and a greater proportion of cases in adults relative to children, consistent with increased population susceptibility. Based on random forest models, A(H1N1) incidence impacts A(H3N2) epidemics to a greater extent than viral evolution, suggesting that subtype interference is a major driver of influenza A virus infection dynamics, presumably via heterosubtypic cross-immunity.
Collapse
Affiliation(s)
- Amanda C Perofsky
- Fogarty International Center, National Institutes of Health, United States
- Brotman Baty Institute for Precision Medicine, University of Washington, United States
| | - John Huddleston
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, United States
| | - Chelsea Hansen
- Fogarty International Center, National Institutes of Health, United States
- Brotman Baty Institute for Precision Medicine, University of Washington, United States
| | - John R Barnes
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), United States
| | - Thomas Rowe
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), United States
| | - Xiyan Xu
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), United States
| | - Rebecca Kondor
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), United States
| | - David E Wentworth
- Virology Surveillance and Diagnosis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), United States
| | - Nicola Lewis
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, United Kingdom
| | - Lynne Whittaker
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, United Kingdom
| | - Burcu Ermetal
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, United Kingdom
| | - Ruth Harvey
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, United Kingdom
| | - Monica Galiano
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, United Kingdom
| | - Rodney Stuart Daniels
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, United Kingdom
| | - John W McCauley
- WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, United Kingdom
| | - Seiichiro Fujisaki
- Influenza Virus Research Center, National Institute of Infectious Diseases, Japan
| | - Kazuya Nakamura
- Influenza Virus Research Center, National Institute of Infectious Diseases, Japan
| | - Noriko Kishida
- Influenza Virus Research Center, National Institute of Infectious Diseases, Japan
| | - Shinji Watanabe
- Influenza Virus Research Center, National Institute of Infectious Diseases, Japan
| | - Hideki Hasegawa
- Influenza Virus Research Center, National Institute of Infectious Diseases, Japan
| | - Sheena G Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Australia
| | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Australia
| | - Florian Krammer
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, United States
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, United States
| | - Trevor Bedford
- Brotman Baty Institute for Precision Medicine, University of Washington, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, United States
- Department of Genome Sciences, University of Washington, United States
- Howard Hughes Medical Institute, Seattle, United States
| | - Cécile Viboud
- Fogarty International Center, National Institutes of Health, United States
| |
Collapse
|
16
|
Raha JR, Kim KH, Bhatnagar N, Liu R, Le CTT, Park BR, Grovenstein P, Pal SS, Ko EJ, Shin CH, Wang BZ, Kang SM. Supplementation of seasonal vaccine with multi-subtype neuraminidase and M2 ectodomain virus-like particle improves protection against homologous and heterologous influenza viruses in aged mice. Antiviral Res 2024; 225:105877. [PMID: 38561077 PMCID: PMC11023748 DOI: 10.1016/j.antiviral.2024.105877] [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: 12/19/2023] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024]
Abstract
The conventional inactivated split seasonal influenza vaccine offers low efficacy, particularly in the elderly and against antigenic variants. Here, to improve the efficacy of seasonal vaccination for the elderly population, we tested whether supplementing seasonal bivalent (H1N1 + H3N2) split (S) vaccine with M2 ectodomain repeat and multi-subtype consensus neuraminidase (NA) proteins (N1 NA + N2 NA + flu B NA) on a virus-like particle (NA-M2e) would induce enhanced cross-protection against different influenza viruses in aged mice. Immunization with split vaccine plus NA-M2e (S + NA-M2e) increased vaccine-specific IgG antibodies towards T-helper type 1 responses and hemagglutination inhibition titers. Aged mice with NA-M2e supplemented vaccination were protected against homologous and heterologous viruses at higher efficacies, as evidenced by preventing weight loss, lowering lung viral loads, inducing broadly cross-protective humoral immunity, and IFN-γ+ CD4 and CD8 T cell responses than those with seasonal vaccine. Overall, this study supports a new strategy of NA-M2e supplemented vaccination to enhance protection against homologous and antigenically different viruses in the elderly.
Collapse
Affiliation(s)
- Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Chau Thuy Tien Le
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Phillip Grovenstein
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Eun-Ju Ko
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, South Korea
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30302, USA.
| |
Collapse
|
17
|
Cunliffe RF, Stirling DC, Razzano I, Murugaiah V, Montomoli E, Kim S, Wane M, Horton H, Caproni LJ, Tregoning JS. Optimizing a linear 'Doggybone' DNA vaccine for influenza virus through the incorporation of DNA targeting sequences and neuraminidase antigen. DISCOVERY IMMUNOLOGY 2024; 3:kyad030. [PMID: 38567290 PMCID: PMC10917164 DOI: 10.1093/discim/kyad030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/08/2023] [Accepted: 01/02/2024] [Indexed: 04/04/2024]
Abstract
Influenza virus represents a challenge for traditional vaccine approaches due to its seasonal changes and potential for zoonotic transmission. Nucleic acid vaccines can overcome some of these challenges, especially through the inclusion of multiple antigens to increase the breadth of response. RNA vaccines were an important part of the response to the COVID-19 pandemic, but for future outbreaks DNA vaccines may have some advantages in terms of stability and manufacturing cost that warrant continuing investigation to fully realize their potential. Here, we investigate influenza virus vaccines made using a closed linear DNA platform, Doggybone™ DNA (dbDNA), produced by a rapid and scalable cell-free method. Influenza vaccines have mostly focussed on Haemagglutinin (HA), but the inclusion of Neuraminidase (NA) may provide additional protection. Here, we explored the potential of including NA in a dbDNA vaccine, looking at DNA optimization, mechanism and breadth of protection. We showed that DNA targeting sequences (DTS) improved immune responses against HA but not NA. We explored whether NA vaccine-induced protection against influenza virus infection was cell-mediated, but depletion of CD8 and NK cells made no impact, suggesting it was antibody-mediated. This is reflected in the restriction of protection to homologous strains of influenza virus. Importantly, we saw that including both HA and NA in a single combined vaccine did not dampen the immune response to either one. Overall, we show that linear dbDNA can induce an immune response against NA, which may offer increased protection in instances of HA mismatch where NA remains more conserved.
Collapse
Affiliation(s)
- Robert F Cunliffe
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - David C Stirling
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Ilaria Razzano
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
- VisMederi srl, Siena, 53100, Italia
| | | | - Emanuele Montomoli
- VisMederi srl, Siena, 53100, Italia
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Sungwon Kim
- Touchlight Genetics Ltd, Hampton, TW12 2ER, UK
| | - Madina Wane
- Touchlight Genetics Ltd, Hampton, TW12 2ER, UK
| | | | | | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| |
Collapse
|
18
|
Wu NC, Ellebedy AH. Targeting neuraminidase: the next frontier for broadly protective influenza vaccines. Trends Immunol 2024; 45:11-19. [PMID: 38103991 PMCID: PMC10841738 DOI: 10.1016/j.it.2023.11.001] [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: 10/24/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 12/19/2023]
Abstract
Current seasonal influenza vaccines, which mainly target hemagglutinin (HA), require annual updates due to the continuous antigenic drift of the influenza virus. Developing an influenza vaccine with increased breadth of protection will have significant public health benefits. The recent discovery of broadly protective antibodies to neuraminidase (NA) has provided important insights into developing a universal influenza vaccine, either by improving seasonal influenza vaccines or designing novel immunogens. However, further in-depth molecular characterizations of NA antibody responses are warranted to fully leverage broadly protective NA antibodies for influenza vaccine designs. Overall, we posit that focusing on NA for influenza vaccine development is synergistic with existing efforts targeting HA, and may represent a cost-effective approach to generating a broadly protective influenza vaccine.
Collapse
Affiliation(s)
- Nicholas C Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
19
|
Zhang X, Ross TM. Anti-neuraminidase immunity in the combat against influenza. Expert Rev Vaccines 2024; 23:474-484. [PMID: 38632930 PMCID: PMC11157429 DOI: 10.1080/14760584.2024.2343689] [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: 12/08/2023] [Accepted: 04/12/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Anti-neuraminidase (NA) immunity correlates with the protection against influenza virus infection in both human and animal models. The aim of this review is to better understand the mechanism of anti-NA immunity, and also to evaluate the approaches on developing NA-based influenza vaccines or enhancing immune responses against NA for current influenza vaccines. AREAS COVERED In this review, the structure of influenza neuraminidase, the contribution of anti-NA immunity to protection, as well as the efforts and challenges of targeting the immune responses to NA were discussed. We also listed some of the newly discovered anti-NA monoclonal antibodies and discussed their contribution in therapeutic as well as the antigen design of a broadly protective NA vaccine. EXPERT OPINION Targeting the immune response to both HA and NA may be critical for achieving the optimal protection since there are different mechanisms of HA and NA elicited protective immunity. Monoclonal antibodies (mAbs) that target the conserved protective lateral face or catalytic sites are effective therapeutics. The epitope discovery using monoclonal antibodies may benefit NA-based vaccine elicited broadly reactive antibody responses. Therefore, the potential for a vaccine that elicits cross-reactive antibodies against neuraminidase is a high priority for next-generation influenza vaccines.
Collapse
Affiliation(s)
- Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
20
|
Do THT, Wheatley AK, Kent SJ, Koutsakos M. Influenza B virus neuraminidase: a potential target for next-generation vaccines? Expert Rev Vaccines 2024; 23:39-48. [PMID: 38037386 DOI: 10.1080/14760584.2023.2290691] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/29/2023] [Indexed: 12/02/2023]
Abstract
INTRODUCTION Influenza B viruses (IBV) cause a significant health and economic burden annually. Due to lower antigenic drift rate, less extensive antigenic diversity, and lack of animal reservoirs, the development of highly effective universal vaccines against IBV might be in reach. Current seasonal influenza vaccines are formulated to induce antibodies against the Hemagglutinin (HA) protein, but their effectiveness is reduced by mismatch between vaccine and circulating strains. AREAS COVERED Given antibodies against the Neuraminidase (NA) have been associated with protection during influenza infection, there is considerable interest in the development of NA-based influenza vaccines. This review summarizes insights into the role of NA-based immunity against IBV and highlights knowledge gaps that should be addressed to inform the design of next-generation influenza B vaccines. We discuss how antibodies recognize broadly cross-reactive epitopes on the NA and the lack of understanding of IBV NA antigenic evolution which would benefit vaccine development in the future. EXPERT OPINION Demonstrating NA antibodies as correlates of protection for IBV in humans would be paramount. Determining the extent of IBV NA antigenic evolution will be informative. Finally, it will be critical to determine optimal strategies for incorporating the appropriate NA antigens in existing clinically approved vaccine formulations.
Collapse
Affiliation(s)
- Thi Hoai Thu Do
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
| |
Collapse
|
21
|
Sun X, Ma H, Wang X, Bao Z, Tang S, Yi C, Sun B. Broadly neutralizing antibodies to combat influenza virus infection. Antiviral Res 2024; 221:105785. [PMID: 38145757 DOI: 10.1016/j.antiviral.2023.105785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
The diversified classification and continuous alteration of influenza viruses underscore for antivirals and vaccines that can counter a broad range of influenza subtypes. Hemagglutinin (HA) and neuraminidase (NA) are two principle viral surface targets for broadly neutralizing antibodies. A series of monoclonal antibodies, targeting HA and NA, have been discovered and characterized with a wide range of neutralizing activity against influenza viruses. Clinical studies have demonstrated the safety and efficacy of some HA stem-targeting antibodies against influenza viruses. Broadly neutralizing antibodies (bnAbs) can serve as both prophylactic and therapeutic agents, as well as play a critical role in identifying antigens and epitopes for the development of universal vaccines. In this review, we described and summarized the latest discoveries and advancements of bnAbs against influenza viruses in both pre- and clinical development. Additionally, we assess whether bnAbs can serve as a viable alternative to vaccination against influenza. Finally, we discussed the rationale behind reverse vaccinology, a structure-guided universal vaccine design strategy that efficiently identifies candidate antigens and conserved epitopes that can be targeted by antibodies.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Hanwen Ma
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xuanjia Wang
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhiheng Bao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shubing Tang
- Department of Investigational New Drug, Shanghai Reinovax Biologics Co., Ltd, Shanghai, 200135, China
| | - Chunyan Yi
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Bing Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
22
|
Fox A. Drift and shape-new insights into human immunity against influenza virus neuraminidase. mBio 2023; 14:e0165423. [PMID: 37933976 PMCID: PMC10746272 DOI: 10.1128/mbio.01654-23] [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] [Indexed: 11/08/2023] Open
Abstract
Influenza virus hemagglutinin mediates infection by binding sialic acids, whereas neuraminidase cleaves sialic acids to release progeny virions. Both are targets of protective antibodies, but influenza vaccine strain selection and antigen dose are based on hemagglutinin alone. Virus characterization using first infection ferret sera indicates that escape from hemagglutination inhibiting (HI) antibodies occurs more frequently and is not coordinated with escape from neuraminidase inhibiting (NI) antibodies. A key question addressed by Daulagala et al. (P. Daulagala, B. R. Mann, K. Leung, E. H. Y. Lau, et al., mBio 14:e00084-23, 2023, https://doi.org/10.1128/mbio.00084-23) is how this translates to humans who encounter multiple influenza viruses throughout life. Their cross-sectional study, using sera from a wide age range of participants and H1N1 viruses spanning 1977-2015, indicates that NI antibodies are more broadly cross-reactive than HI antibodies. Both HI and NI titers were highest against strains encountered in childhood indicating that both are shaped by priming exposures. The study further supports the development of NA-optimized vaccines.
Collapse
Affiliation(s)
- Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
McMahon M, Tan J, O'Dell G, Kirkpatrick Roubidoux E, Strohmeier S, Krammer F. Immunity induced by vaccination with recombinant influenza B virus neuraminidase protein breaks viral transmission chains in guinea pigs in an exposure intensity-dependent manner. J Virol 2023; 97:e0105723. [PMID: 37800945 PMCID: PMC10617433 DOI: 10.1128/jvi.01057-23] [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: 07/14/2023] [Accepted: 08/22/2023] [Indexed: 10/07/2023] Open
Abstract
IMPORTANCE Vaccines that can slow respiratory virus transmission in the population are urgently needed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus. Here, we describe how a recombinant neuraminidase-based influenza virus vaccine reduces transmission in vaccinated guinea pigs in an exposure intensity-based manner.
Collapse
Affiliation(s)
- Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jessica Tan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - George O'Dell
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ericka Kirkpatrick Roubidoux
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
24
|
Cheung M, Chang C, Rathnasinghe R, Rossignol E, Zhang Y, Ferrari A, Patel H, Huang Y, Sanchez Guillen M, Scalzo T, Lee C, Otten GR, Settembre EC, Music N, Palladino G, Wen Y. Self-amplifying mRNA seasonal influenza vaccines elicit mouse neutralizing antibody and cell-mediated immunity and protect ferrets. NPJ Vaccines 2023; 8:150. [PMID: 37794051 PMCID: PMC10550923 DOI: 10.1038/s41541-023-00747-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023] Open
Abstract
Currently licensed influenza vaccines focus immune responses on viral hemagglutinin (HA), while the other major surface glycoprotein neuraminidase (NA) is not tightly controlled in inactivated vaccine formulations despite evidence that anti-NA antibodies reduce clinical disease. We utilized a bicistronic self-amplifying mRNA (sa-mRNA) platform encoding both HA and NA from four seasonal influenza strains, creating a quadrivalent influenza vaccine. sa-mRNA vaccines encoding an NA component induced the production of NA-inhibiting antibodies and CD4+ T-cell responses in both monovalent and quadrivalent formulations. Including NA in the vaccine enabled cross-neutralization against antigenically drifted strains and provided greater protection than HA alone upon A(H3N2) challenge in ferrets. These results demonstrate that next-generation bicistronic sa-mRNA vaccines expressing HA and NA induce potent antibodies against both viral coat proteins, as well as vaccine-specific cell-mediated immunity. When formulated as a quadrivalent seasonal influenza vaccine, the sa-mRNA platform provides an opportunity to increase the breadth of protection through cross-neutralizing anti-NA antibodies.
Collapse
Affiliation(s)
| | - Cheng Chang
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | | | - Yunfei Zhang
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | - Harsh Patel
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | - Yanjun Huang
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | - Tina Scalzo
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | - Changkeun Lee
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | | | - Nedzad Music
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | - Yingxia Wen
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA.
| |
Collapse
|
25
|
Puente-Massaguer E, Vasilev K, Beyer A, Loganathan M, Francis B, Scherm MJ, Arunkumar GA, González-Domínguez I, Zhu X, Wilson IA, Coughlan L, Sun W, Palese P, Krammer F. Chimeric hemagglutinin split vaccines elicit broadly cross-reactive antibodies and protection against group 2 influenza viruses in mice. SCIENCE ADVANCES 2023; 9:eadi4753. [PMID: 37703367 PMCID: PMC10499326 DOI: 10.1126/sciadv.adi4753] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023]
Abstract
Seasonal influenza virus vaccines are effective when they are well matched to circulating strains. Because of antigenic drift/change in the immunodominant hemagglutinin (HA) head domain, annual vaccine reformulations are necessary to maintain a match with circulating strains. In addition, seasonal vaccines provide little to no protection against newly emerging pandemic strains. Sequential vaccination with chimeric HA (cHA) constructs has been proven to direct the immune response toward the immunosubdominant but more conserved HA stalk domain. In this study, we show that immunization with group 2 cHA split vaccines in combination with the CpG 1018 adjuvant elicits broadly cross-reactive antibodies against all group 2 HAs, as well as systemic and local antigen-specific T cell responses. Antibodies elicited after sequential vaccination are directed to conserved regions of the HA such as the stalk and the trimer interface and also to the N2 neuraminidase (NA). Immunized mice were fully protected from challenge with a broad panel of influenza A viruses.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kirill Vasilev
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Annika Beyer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Benjamin Francis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael J. Scherm
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | | | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
26
|
Liu X, Zhao T, Wang L, Yang Z, Luo C, Li M, Luo H, Sun C, Yan H, Shu Y. A mosaic influenza virus-like particles vaccine provides broad humoral and cellular immune responses against influenza A viruses. NPJ Vaccines 2023; 8:132. [PMID: 37679361 PMCID: PMC10485063 DOI: 10.1038/s41541-023-00728-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The development of a universal influenza vaccine to elicit broad immune responses is essential in reducing disease burden and pandemic impact. In this study, the mosaic vaccine design strategy and genetic algorithms were utilized to optimize the seasonal influenza A virus (H1N1, H3N2) hemagglutinin (HA) and neuraminidase (NA) antigens, which also contain most potential T-cell epitopes. These mosaic immunogens were then expressed as virus-like particles (VLPs) using the baculovirus expression system. The immunogenicity and protection effectiveness of the mosaic VLPs were compared to the commercial quadrivalent inactivated influenza vaccine (QIV) in the mice model. Strong cross-reactive antibody responses were observed in mice following two doses of vaccination with the mosaic VLPs, with HI titers higher than 40 in 15 of 16 tested strains as opposed to limited cross HI antibody levels with QIV vaccination. After a single vaccination, mice also show a stronger level of cross-reactive antibody responses than the QIV. The QIV vaccinations only elicited NI antibodies to a small number of vaccine strains, and not even strong NI antibodies to its corresponding vaccine components. In contrast, the mosaic VLPs caused robust NI antibodies to all tested seasonal influenza virus vaccine strains. Here, we demonstrated the mosaic vaccines induces stronger cross-reactive antibodies and robust more T-cell responses compared to the QIV. The mosaic VLPs also provided protection against challenges with ancestral influenza A viruses of both H1 and H3 subtypes. These findings indicated that the mosaic VLPs were a promising strategy for developing a broad influenza vaccine in future.
Collapse
Affiliation(s)
- Xuejie Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Tianyi Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Liangliang Wang
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Zhuolin Yang
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Chuming Luo
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Minchao Li
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Huanle Luo
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Caijun Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China.
| | - Huacheng Yan
- Center for Disease Control and Prevention of Southern Military Theatre, 510610, Guangzhou, China.
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China.
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China.
| |
Collapse
|
27
|
Mettelman RC, Souquette A, Van de Velde LA, Vegesana K, Allen EK, Kackos CM, Trifkovic S, DeBeauchamp J, Wilson TL, St James DG, Menon SS, Wood T, Jelley L, Webby RJ, Huang QS, Thomas PG. Baseline innate and T cell populations are correlates of protection against symptomatic influenza virus infection independent of serology. Nat Immunol 2023; 24:1511-1526. [PMID: 37592015 PMCID: PMC10566627 DOI: 10.1038/s41590-023-01590-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Evidence suggests that innate and adaptive cellular responses mediate resistance to the influenza virus and confer protection after vaccination. However, few studies have resolved the contribution of cellular responses within the context of preexisting antibody titers. Here, we measured the peripheral immune profiles of 206 vaccinated or unvaccinated adults to determine how baseline variations in the cellular and humoral immune compartments contribute independently or synergistically to the risk of developing symptomatic influenza. Protection correlated with diverse and polyfunctional CD4+ and CD8+ T, circulating T follicular helper, T helper type 17, myeloid dendritic and CD16+ natural killer (NK) cell subsets. Conversely, increased susceptibility was predominantly attributed to nonspecific inflammatory populations, including γδ T cells and activated CD16- NK cells, as well as TNFα+ single-cytokine-producing CD8+ T cells. Multivariate and predictive modeling indicated that cellular subsets (1) work synergistically with humoral immunity to confer protection, (2) improve model performance over demographic and serologic factors alone and (3) comprise the most important predictive covariates. Together, these results demonstrate that preinfection peripheral cell composition improves the prediction of symptomatic influenza susceptibility over vaccination, demographics or serology alone.
Collapse
Affiliation(s)
- Robert C Mettelman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Aisha Souquette
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lee-Ann Van de Velde
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kasi Vegesana
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - E Kaitlynn Allen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christina M Kackos
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sanja Trifkovic
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jennifer DeBeauchamp
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taylor L Wilson
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Microbiology, Immunology and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Deryn G St James
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Microbiology, Immunology and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Smrithi S Menon
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Timothy Wood
- Institute of Environmental Science and Research Limited (ESR), Wallaceville Science Centre, Upper Hutt, New Zealand
| | - Lauren Jelley
- Institute of Environmental Science and Research Limited (ESR), Wallaceville Science Centre, Upper Hutt, New Zealand
| | - Richard J Webby
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Q Sue Huang
- Institute of Environmental Science and Research Limited (ESR), Wallaceville Science Centre, Upper Hutt, New Zealand.
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
28
|
Boudreau CM, Burke JS, Yousif AS, Sangesland M, Jastrzebski S, Verschoor C, Kuchel G, Lingwood D, Kleanthous H, De Bruijn I, Landolfi V, Sridhar S, Alter G. Antibody-mediated NK cell activation as a correlate of immunity against influenza infection. Nat Commun 2023; 14:5170. [PMID: 37620306 PMCID: PMC10449820 DOI: 10.1038/s41467-023-40699-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Antibodies play a critical role in protection against influenza; yet titers and viral neutralization represent incomplete correlates of immunity. Instead, the ability of antibodies to leverage the antiviral power of the innate immune system has been implicated in protection from and clearance of influenza infection. Here, post-hoc analysis of the humoral immune response to influenza is comprehensively profiled in a cohort of vaccinated older adults (65 + ) monitored for influenza infection during the 2012/2013 season in the United States (NCT: 01427309). While robust humoral immune responses arose against the vaccine and circulating strains, influenza-specific antibody effector profiles differed in individuals that later became infected with influenza, who are deficient in NK cell activating antibodies to both hemagglutinin and neuraminidase, compared to individuals who remained uninfected. Furthermore, NK cell activation was strongly associated with the NK cell senescence marker CD57, arguing for the need for selective induction of influenza-specific afucosylated NK activating antibodies in older adults to achieve protection. High dose vaccination, currently used for older adults, was insufficient to generate this NK cell-activating humoral response. Next generation vaccines able to selectively bolster NK cell activating antibodies may be required to achieve protection in the setting of progressively senescent NK cells.
Collapse
Affiliation(s)
- Carolyn M Boudreau
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02129, USA
- PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA, 02115, USA
| | - John S Burke
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02129, USA
| | - Ashraf S Yousif
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02129, USA
| | - Maya Sangesland
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02129, USA
- PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA, 02115, USA
| | | | - Chris Verschoor
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - George Kuchel
- Center on Aging, UCONN Health Center, Farmington, CT, 06030, USA
| | - Daniel Lingwood
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02129, USA
| | | | | | | | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02129, USA.
| |
Collapse
|
29
|
Hansen L, McMahon M, Turner HL, Zhu X, Turner JS, Ozorowski G, Stadlbauer D, Vahokoski J, Schmitz AJ, Rizk AA, Alsoussi WB, Strohmeier S, Yu W, Choreño-Parra JA, Jiménez-Alvarez L, Cruz-Lagunas A, Zúñiga J, Mudd PA, Cox RJ, Wilson IA, Ward AB, Ellebedy AH, Krammer F. Human anti-N1 monoclonal antibodies elicited by pandemic H1N1 virus infection broadly inhibit HxN1 viruses in vitro and in vivo. Immunity 2023; 56:1927-1938.e8. [PMID: 37506693 PMCID: PMC10529248 DOI: 10.1016/j.immuni.2023.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
Neuraminidase (NA) is one of the two influenza virus surface glycoproteins, and antibodies that target it are an independent correlate of protection. However, our current understanding of NA antigenicity is incomplete. Here, we describe human monoclonal antibodies (mAbs) from a patient with a pandemic H1N1 virus infection in 2009. Two mAbs exhibited broad reactivity and inhibited NA enzyme activity of seasonal H1N1 viruses circulating before and after 2009, as well as viruses with avian or swine N1s. The mAbs provided robust protection from lethal challenge with human H1N1 and avian H5N1 viruses in mice, and both target an epitope on the lateral face of NA. In summary, we identified two broadly protective NA antibodies that share a novel epitope, inhibited NA activity, and provide protection against virus challenge in mice. Our work reaffirms that NA should be included as a target in future broadly protective or universal influenza virus vaccines.
Collapse
Affiliation(s)
- Lena Hansen
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hannah L Turner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juha Vahokoski
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway; National Advisory Unit for Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| | - Aaron J Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Amena A Rizk
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wafaa B Alsoussi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenli Yu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - José Alberto Choreño-Parra
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Luis Jiménez-Alvarez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico; Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Philip A Mudd
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rebecca J Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
30
|
Werninghaus IC, Hinke DM, Fossum E, Bogen B, Braathen R. Neuraminidase delivered as an APC-targeted DNA vaccine induces protective antibodies against influenza. Mol Ther 2023; 31:2188-2205. [PMID: 36926694 PMCID: PMC10362400 DOI: 10.1016/j.ymthe.2023.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 02/01/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Conventional influenza vaccines focus on hemagglutinin (HA). However, antibody responses to neuraminidase (NA) have been established as an independent correlate of protection. Here, we introduced the ectodomain of NA into DNA vaccines that, as translated dimeric vaccine proteins, target antigen-presenting cells (APCs). The targeting was mediated by an single-chain variable fragment specific for major histocompatibility complex (MHC) class II, which is genetically linked to NA via a dimerization motif. A single immunization of BALB/c mice elicited strong and long-lasting NA-specific antibodies that inhibited NA enzymatic activity and reduced viral replication. Vaccine-induced NA immunity completely protected against a homologous influenza virus and out-competed NA immunity induced by a conventional inactivated virus vaccine. The protection was mainly mediated by antibodies, although NA-specific T cells also contributed. APC-targeting and antigen bivalency were crucial for vaccine efficacy. The APC-targeted vaccine was potent at low doses of DNA, indicating a dose-sparing effect. Similar results were obtained with NA vaccines that targeted different surface molecules on dendritic cells. Interestingly, the protective efficacy of NA as antigen compared favorably with HA and therefore ought to receive more attention in influenza vaccine research.
Collapse
Affiliation(s)
- Ina Charlotta Werninghaus
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway; Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, 0372 Oslo, Norway.
| | - Daniëla Maria Hinke
- Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, 0372 Oslo, Norway
| | - Even Fossum
- Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, 0372 Oslo, Norway
| | - Bjarne Bogen
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway; Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, 0372 Oslo, Norway
| | - Ranveig Braathen
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway; Division of Laboratory Medicine, Department of Immunology, Oslo University Hospital, 0372 Oslo, Norway.
| |
Collapse
|
31
|
Martinez MR, Gao J, Wan H, Kang H, Klenow L, Daniels R. Inactivated influenza virions are a flexible vaccine platform for eliciting protective antibody responses against neuraminidase. Vaccine 2023:S0264-410X(23)00629-1. [PMID: 37301705 DOI: 10.1016/j.vaccine.2023.05.068] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
Most seasonal influenza vaccines are produced using hemagglutinin (HA) surface antigens from inactivated virions. However, virions are thought to be a suboptimal source for the less abundant neuraminidase (NA) surface antigen, which is also protective against severe disease. Here, we demonstrate that inactivated influenza virions are compatible with two modern approaches for improving protective antibody responses against NA. Using a DBA/2J mouse model, we show that the strong infection-induced NA inhibitory (NAI) antibody responses are only achieved by high dose immunizations of inactivated virions, likely due to the low viral NA content. Based on this observation, we first produced virions with higher NA content by using reverse genetics to exchange the viral internal gene segments. Single immunizations with these inactivated virions showed enhanced NAI antibody responses and improved NA-based protection from a lethal viral challenge while also allowing for the development of natural immunity to the heterotypic challenge virus HA. Second, we combined inactivated virions with recombinant NA protein antigens. These combination vaccines increased NA-based protection following viral challenge and elicited stronger antibody responses against NA than either component alone, especially when the NAs possessed similar antigenicity. Together, these results indicate that inactivated virions are a flexible platform that can be easily combined with protein-based vaccines to improve protective antibody responses against influenza antigens.
Collapse
Affiliation(s)
- Mira Rakic Martinez
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Jin Gao
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hongquan Wan
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hyeog Kang
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Robert Daniels
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
32
|
Oishi K, Blanco-Melo D, Kurland AP, Johnson JR, tenOever BR. Archaeal Kink-Turn Binding Protein Mediates Inhibition of Orthomyxovirus Splicing Biology. J Virol 2023; 97:e0181322. [PMID: 36943134 PMCID: PMC10134859 DOI: 10.1128/jvi.01813-22] [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: 11/22/2022] [Accepted: 02/22/2023] [Indexed: 03/23/2023] Open
Abstract
Despite lacking a DNA intermediate, orthomyxoviruses complete their replication cycle in the nucleus and generate multiple transcripts by usurping the host splicing machinery. This biology results in dynamic changes of relative viral transcripts over time and dictates the replicative phase of the infection. Here, we demonstrate that the family of archaeal L7Ae proteins uniquely inhibit the splicing biology of influenza A virus, influenza B virus, and Salmon isavirus, revealing a common strategy utilized by Orthomyxoviridae members to achieve this dynamic. L7Ae-mediated inhibition of virus biology was lost with the generation of a splicing-independent strain of influenza A virus and attempts to select for an escape mutant resulted in variants that conformed to host splicing biology at significant cost to their overall fitness. As L7Ae recognizes conventional kink turns in various RNAs, these data implicate the formation of a similar structure as a shared strategy adopted by this virus family to coordinate their replication cycle. IMPORTANCE Here, we demonstrate that a family of proteins from archaea specifically inhibit this splicing biology of all tested members of the Orthomyxoviridae family. We show that this inhibition extends to influenza A virus, influenza B virus, and isavirus genera, while having no significant impact on the mammalian transcriptome or proteome. Attempts to generate an escape mutant against L7Ae-mediated inhibition resulted in mutations surrounding the viral splice sites and a significant loss of viral fitness. Together, these findings reveal a unique biology shared among diverse members of the Orthomyxoviridae family that may serve as a means to generate future universal therapeutics.
Collapse
Affiliation(s)
- Kohei Oishi
- Department of Microbiology, New York University, Grossman School of Medicine, New York, New York, USA
| | - Daniel Blanco-Melo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andrew P. Kurland
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jeffrey R. Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benjamin R. tenOever
- Department of Microbiology, New York University, Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
33
|
Amanat F, Clark J, Carreño JM, Strohmeier S, Yellin T, Meade PS, Bhavsar D, Muramatsu H, Sun W, Coughlan L, Pardi N, Krammer F. Immunity to Seasonal Coronavirus Spike Proteins Does Not Protect from SARS-CoV-2 Challenge in a Mouse Model but Has No Detrimental Effect on Protection Mediated by COVID-19 mRNA Vaccination. J Virol 2023; 97:e0166422. [PMID: 36779758 PMCID: PMC10062180 DOI: 10.1128/jvi.01664-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/14/2023] [Indexed: 02/14/2023] Open
Abstract
Seasonal coronaviruses have been circulating widely in the human population for many years. With increasing age, humans are more likely to have been exposed to these viruses and to have developed immunity against them. It has been hypothesized that this immunity to seasonal coronaviruses may provide partial protection against infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and it has also been shown that coronavirus disease 2019 (COVID-19) vaccination induces a back-boosting effects against the spike proteins of seasonal betacoronaviruses. In this study, we tested if immunity to the seasonal coronavirus spikes from OC43, HKU1, 229E, or NL63 would confer protection against SARS-CoV-2 challenge in a mouse model, and whether pre-existing immunity against these spikes would weaken the protection afforded by mRNA COVID-19 vaccination. We found that mice vaccinated with the seasonal coronavirus spike proteins had no increased protection compared to the negative controls. While a negligible back-boosting effect against betacoronavirus spike proteins was observed after SARS-CoV-2 infection, there was no negative original antigenic sin-like effect on the immune response and protection induced by SARS-CoV-2 mRNA vaccination in animals with pre-existing immunity to seasonal coronavirus spike proteins. IMPORTANCE The impact that immunity against seasonal coronaviruses has on both susceptibility to SARS-CoV-2 infection as well as on COVID-19 vaccination is unclear. This study provides insights into both questions in a mouse model of SARS-CoV-2.
Collapse
Affiliation(s)
- Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jordan Clark
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Temima Yellin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Philip S. Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Disha Bhavsar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
34
|
Cable J, Balachandran S, Daley-Bauer LP, Rustagi A, Antony F, Frere JJ, Strampe J, Kedzierska K, Cannon JL, McGargill MA, Weiskopf D, Mettelman RC, Niessl J, Thomas PG, Briney B, Valkenburg SA, Bloom JD, Bjorkman PJ, Iketani S, Rappazzo CG, Crooks CM, Crofts KF, Pöhlmann S, Krammer F, Sant AJ, Nabel GJ, Schultz-Cherry S. Viral immunity: Basic mechanisms and therapeutic applications-a Keystone Symposia report. Ann N Y Acad Sci 2023; 1521:32-45. [PMID: 36718537 DOI: 10.1111/nyas.14960] [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] [Indexed: 02/01/2023]
Abstract
Viruses infect millions of people each year. Both endemic viruses circulating throughout the population as well as novel epidemic and pandemic viruses pose ongoing threats to global public health. Developing more effective tools to address viruses requires not only in-depth knowledge of the virus itself but also of our immune system's response to infection. On June 29 to July 2, 2022, researchers met for the Keystone symposium "Viral Immunity: Basic Mechanisms and Therapeutic Applications." This report presents concise summaries from several of the symposium presenters.
Collapse
Affiliation(s)
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Lisa P Daley-Bauer
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arjun Rustagi
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ferrin Antony
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Justin J Frere
- East Harlem Health Outreach Partnership; Department of Medical Education; and Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jamie Strampe
- Bioinformatics Program, Boston University and National Emerging Infectious Diseases Laboratories, Boston, Massachusetts, USA
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Judy L Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Maureen A McGargill
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, California, USA
| | - Robert C Mettelman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Julia Niessl
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Bryan Briney
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, PR China
| | - Jesse D Bloom
- Basic Sciences Division and Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Microbiology and Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Sho Iketani
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | | - Chelsea M Crooks
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kali F Crofts
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center and Faculty of Biology and Psychology, University Medical Center Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrea J Sant
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gary J Nabel
- Modex Therapeutics Inc., an OPKO Health Company, Natick, Massachusetts, USA
| | - Stacey Schultz-Cherry
- Department of Laboratory Medicine and Department of Immunology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
35
|
Hufnagel DE, Young KM, Arendsee ZW, Gay LC, Caceres CJ, Rajão DS, Perez DR, Vincent Baker AL, Anderson TK. Characterizing a century of genetic diversity and contemporary antigenic diversity of N1 neuraminidase in influenza A virus from North American swine. Virus Evol 2023; 9:vead015. [PMID: 36993794 PMCID: PMC10041950 DOI: 10.1093/ve/vead015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/08/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Influenza A viruses (IAVs) of the H1N1 classical swine lineage became endemic in North American swine following the 1918 pandemic. Additional human-to-swine transmission events after 1918, and a spillover of H1 viruses from wild birds in Europe, potentiated a rapid increase in genomic diversity via reassortment between introductions and the endemic classical swine lineage. To determine mechanisms affecting reassortment and evolution, we conducted a phylogenetic analysis of N1 and paired HA swine IAV genes in North America between 1930 and 2020. We described fourteen N1 clades within the N1 Eurasian avian lineage (including the N1 pandemic clade), the N1 classical swine lineage, and the N1 human seasonal lineage. Seven N1 genetic clades had evidence for contemporary circulation. To assess antigenic drift associated with N1 genetic diversity, we generated a panel of representative swine N1 antisera and quantified the antigenic distance between wild-type viruses using enzyme-linked lectin assays and antigenic cartography. Within the N1 genes, the antigenic similarity was variable and reflected shared evolutionary history. Sustained circulation and evolution of N1 genes in swine had resulted in a significant antigenic distance between the N1 pandemic clade and the classical swine lineage. Between 2010 and 2020, N1 clades and N1-HA pairings fluctuated in detection frequency across North America, with hotspots of diversity generally appearing and disappearing within 2 years. We also identified frequent N1-HA reassortment events (n = 36), which were rarely sustained (n = 6) and sometimes also concomitant with the emergence of new N1 genetic clades (n = 3). These data form a baseline from which we can identify N1 clades that expand in range or genetic diversity that may impact viral phenotypes or vaccine immunity and subsequently the health of North American swine.
Collapse
Affiliation(s)
- David E Hufnagel
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Katharine M Young
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Zebulun W Arendsee
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA 50010, USA
| | - L Claire Gay
- Department of Population Health, College of Veterinary Medicine, University of Georgia, 953 College Station Rd, Athens, GA 30602, USA
| | - C Joaquin Caceres
- Department of Population Health, College of Veterinary Medicine, University of Georgia, 953 College Station Rd, Athens, GA 30602, USA
| | - Daniela S Rajão
- Department of Population Health, College of Veterinary Medicine, University of Georgia, 953 College Station Rd, Athens, GA 30602, USA
| | - Daniel R Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, 953 College Station Rd, Athens, GA 30602, USA
| | - Amy L Vincent Baker
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Tavis K Anderson
- Virus and Prion Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA 50010, USA
| |
Collapse
|
36
|
Designing multi-epitope mRNA construct as a universal influenza vaccine candidate for future epidemic/pandemic preparedness. Int J Biol Macromol 2023; 226:885-899. [PMID: 36521707 DOI: 10.1016/j.ijbiomac.2022.12.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/25/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Despite the availability of prevention and treatment strategies and advancing immunization approaches, the influenza virus remains a global threat that continues to plague humanity with unpredictable pandemics. Due to the unusual genetic variability and segmented genome, the reassortment between different strains of influenza is facilitated and the viruses continuously evolve and adapt to the host cell's immunity. This underlies the seasonal vaccine mismatches that decrease the vaccine efficacy and increase the risk of outbreaks. Thus, the development of a universal vaccine covering all the influenza A and B strains would reduce the pervasiveness of the influenza virus. In the current study, a potentially universal influenza multi-epitope vaccine was designed based on the experimentally tested conserved T cell and B cell epitopes of hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), and matrix-2 proton channel (M2) of the virus. The immune simulation and molecular docking of the vaccine construct with TLR2, TLR3, and TLR4 elicited the favorable immunogenicity of the vaccine and the formation of stable complexes, respectively. Ultimately, based on the immunoinformatics analysis, the universal mRNA multi-epitope vaccine designed in this study might have a protection potential against the various subtypes of influenza A and B.
Collapse
|
37
|
Abbadi N, Mousa JJ. Broadly Protective Neuraminidase-Based Influenza Vaccines and Monoclonal Antibodies: Target Epitopes and Mechanisms of Action. Viruses 2023; 15:200. [PMID: 36680239 PMCID: PMC9861061 DOI: 10.3390/v15010200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Neuraminidase (NA) is an important surface protein on influenza virions, playing an essential role in the viral life cycle and being a key target of the immune system. Despite the importance of NA-based immunity, current vaccines are focused on the hemagglutinin (HA) protein as the target for protective antibodies, and the amount of NA is not standardized in virion-based vaccines. Antibodies targeting NA are predominantly protective, reducing infection severity and viral shedding. Recently, NA-specific monoclonal antibodies have been characterized, and their target epitopes have been identified. This review summarizes the characteristics of NA, NA-specific antibodies, the mechanism of NA inhibition, and the recent efforts towards developing NA-based and NA-incorporating influenza vaccines.
Collapse
Affiliation(s)
- Nada Abbadi
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Jarrod J. Mousa
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
38
|
Skarlupka AL, Zhang X, Blas-Machado U, Sumner SF, Ross TM. Multi-Influenza HA Subtype Protection of Ferrets Vaccinated with an N1 COBRA-Based Neuraminidase. Viruses 2023; 15:184. [PMID: 36680224 PMCID: PMC9865009 DOI: 10.3390/v15010184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/25/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
The influenza neuraminidase (NA) is a promising target for next-generation vaccines. Protection induced by vaccination with the computationally optimized broadly reactive NA antigen (N1-I COBRA NA) was characterized in both influenza serologically naive and pre-immune ferret models following H1N1 (A/California/07/2009, CA/09) or H5N1 (A/Vietnam/1203/2004, Viet/04) influenza challenges. The N1-I COBRA NA vaccine elicited antibodies with neutralizing ELLA activity against both seasonal and pandemic H1N1 influenza, as well as the H5N1 influenza virus. In both models, N1-I COBRA NA-vaccinated ferrets that were challenged with CA/09 virus had similar morbidity (weight loss and clinical symptoms) as ferrets vaccinated with the CA/09 HA control vaccine. There were significantly reduced viral titers compared to the mock-vaccinated control animals. Ferrets vaccinated with N1-I COBRA NA or Viet/04 NA vaccines were protected against the H5N1 virus infection with minimal clinical symptoms and negligible weight loss. In contrast, ferrets vaccinated with the CA/09 NA vaccine lost ~10% of their original body weight with 25% mortality. Vaccination with either HA or NA vaccines did not inhibit contact transmission of CA/09 virus to naïve cage mates. Overall, the N1-I COBRA vaccine elicited protective immune responses against both H1N1 and H5N1 infections and partially mitigated disease in contact-transmission receiving ferrets. These results indicate that the N1-I COBRA NA performed similarly to the CA/09 HA and NA positive controls. Therefore, the N1-I COBRA NA alone induces protection against viruses from both H5N1 and H1N1 subtypes, indicating its value as a vaccine component in broadly protective influenza vaccines.
Collapse
Affiliation(s)
- Amanda L. Skarlupka
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Uriel Blas-Machado
- Athens Veterinary Diagnostic Laboratory, University of Georgia, Athens, GA 30602, USA
- Department of Pathology, University of Georgia, Athens, GA 30602, USA
| | - Spencer F. Sumner
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| |
Collapse
|
39
|
Yasuhara A, Yamayoshi S, Kiso M, Sakai-Tagawa Y, Okuda M, Kawaoka Y. A broadly protective human monoclonal antibody targeting the sialidase activity of influenza A and B virus neuraminidases. Nat Commun 2022; 13:6602. [PMID: 36329075 PMCID: PMC9632566 DOI: 10.1038/s41467-022-34521-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Improved vaccines and antiviral agents that provide better, broader protection against seasonal and emerging influenza viruses are needed. The viral surface glycoprotein hemagglutinin (HA) is a primary target for the development of universal influenza vaccines and therapeutic antibodies. The other major surface antigen, neuraminidase (NA), has been less well studied as a potential target and fewer broadly reactive anti-NA antibodies have been identified. In this study, we isolate three human monoclonal antibodies that recognize NA from A/H1N1 subtypes, and find that one of them, clone DA03E17, binds to the NA of A/H3N2, A/H5N1, A/H7N9, B/Ancestral-lineage, B/Yamagata-lineage, and B/Victoria-lineage viruses. DA03E17 inhibits the neuraminidase activity by direct binding to the enzyme active site, and provides in vitro and in vivo protection against infection with several types of influenza virus. This clone could, therefore, be useful as a broadly protective therapeutic agent. Moreover, the neutralizing epitope of DA03E17 could be useful in the development of an NA-based universal influenza vaccine.
Collapse
Affiliation(s)
- Atsuhiro Yasuhara
- grid.26999.3d0000 0001 2151 536XDivision of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Yamayoshi
- grid.26999.3d0000 0001 2151 536XDivision of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XInternational Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan ,grid.45203.300000 0004 0489 0290Research Center for Global Viral Infections, National Center for Global Health and Medicine, Tokyo, Japan
| | - Maki Kiso
- grid.26999.3d0000 0001 2151 536XDivision of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuko Sakai-Tagawa
- grid.26999.3d0000 0001 2151 536XDivision of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Moe Okuda
- grid.26999.3d0000 0001 2151 536XDivision of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Kawaoka
- grid.26999.3d0000 0001 2151 536XDivision of Virology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan ,grid.26999.3d0000 0001 2151 536XInternational Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan ,grid.45203.300000 0004 0489 0290Research Center for Global Viral Infections, National Center for Global Health and Medicine, Tokyo, Japan ,grid.14003.360000 0001 2167 3675Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI USA
| |
Collapse
|
40
|
McMahon M, Tan J, O'Dell G, Roubidoux EK, Strohmeier S, Krammer F. Immunity induced by vaccination with recombinant influenza B virus neuraminidase protein breaks viral transmission chains in guinea pigs in an exposure intensity-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.10.19.512980. [PMID: 36299418 PMCID: PMC9603823 DOI: 10.1101/2022.10.19.512980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mucosal vaccines and vaccines that block pathogen transmission are under-appreciated in vaccine development. However, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has shown that blocking viral transmission is an important attribute of efficient vaccines. Here, we investigated if recombinant influenza virus neuraminidase (NA) vaccines delivered at a mucosal site could protect from onward transmission of influenza B viruses in the guinea pig model. We tested four different scenarios in which sequential transmission was investigated in chains of four guinea pigs. The variables tested included a low and a high viral inoculum (104 vs 105 plaque forming units) in the initial donor guinea pig and variation of exposure/cohousing time (1 day vs 6 days). In three out of four scenarios - low inoculum-long exposure, low inoculum-short exposure and high inoculum-short exposure - transmission chains were efficiently blocked. Based on this data we believe an intranasal recombinant NA vaccine could be used to efficiently curtail influenza virus spread in the human population during influenza epidemics.
Collapse
Affiliation(s)
- Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Tan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George O'Dell
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ericka Kirkpatrick Roubidoux
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
41
|
Sicca F, Sakorafa E, de Jonge A, de Vries-Idema J, Zhou F, Cox RJ, Huckriede A. The evolution of humoral immune responses to past and novel influenza virus strains gives evidence for antigenic seniority. Front Immunol 2022; 13:987984. [PMID: 36119111 PMCID: PMC9478913 DOI: 10.3389/fimmu.2022.987984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022] Open
Abstract
The high genetic and antigenic variability of influenza virus and the repeated exposures of individuals to the virus over time account for the human immune responses toward this pathogen to continuously evolve during the lifespan of an individual. Influenza-specific immune memory to past strains has been shown to affect the immune responses to subsequent influenza strains and in turn to be changed itself through the new virus encounter. However, exactly how and to what extent this happens remains unclear. Here we studied pre-existing immunity against influenza A virus (IAV) by assessing IAV binding (IgG), neutralizing, and neuraminidase-specific antibodies to 5 different IAV strains in 180 subjects from 3 different age cohorts, adolescents, adults, and elderly, over a 5-year time span. In each age cohort, the highest neutralizing antibody titers were seen for a virus strain that circulated early in their life but the highest increase in titer was found for the most recent virus strains. In contrast, the highest IgG titers were seen against recent virus strains but the biggest increase in titer occurred against older strains. Significant increases in neutralizing antibody titers against a newly encountered virus strain were observed in all age cohorts demonstrating that pre-existing immunity did not hamper antibody induction. Our results indicate that the evolution of influenza-specific humoral immunity differs for rather cross-reactive virus-binding antibodies and more strain-specific neutralizing antibodies. Nevertheless, in general, our observations lend support to the antigenic seniority theory according to which the antibody response to influenza is broadened with each virus encounter, with the earliest encountered strain taking in the most senior and thus dominant position.
Collapse
Affiliation(s)
- Federica Sicca
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Eleni Sakorafa
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Anouk de Jonge
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jacqueline de Vries-Idema
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Rebecca Jane Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Anke Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- *Correspondence: Anke Huckriede,
| |
Collapse
|
42
|
Development of a pentavalent broadly protective nucleoside-modified mRNA vaccine against influenza B viruses. Nat Commun 2022; 13:4677. [PMID: 35945226 PMCID: PMC9362976 DOI: 10.1038/s41467-022-32149-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 07/20/2022] [Indexed: 11/09/2022] Open
Abstract
Messenger RNA (mRNA) vaccines represent a new, effective vaccine platform with high capacity for rapid development. Generation of a universal influenza virus vaccine with the potential to elicit long-lasting, broadly cross-reactive immune responses is a necessity for reducing influenza-associated morbidity and mortality. Here we focus on the development of a universal influenza B virus vaccine based on the lipid nanoparticle-encapsulated nucleoside-modified mRNA (mRNA-LNP) platform. We evaluate vaccine candidates based on different target antigens that afford protection against challenge with ancestral and recent influenza B viruses from both antigenic lineages. A pentavalent vaccine combining all tested antigens protects mice from morbidity at a very low dose of 50 ng per antigen after a single vaccination. These findings support the further advancement of nucleoside-modified mRNA-LNPs expressing multiple conserved antigens as universal influenza virus vaccine candidates. The public health concern caused by influenza B virus is often overlooked, yet represents a significant global burden. Here, the authors evaluate the cellular and humoral immune responses of multivalent vaccine candidates, based on the lipid nanoparticle-encapsulated nucleoside-modified mRNA platform, and demonstrate protection of mice from challenge with a broad panel of influenza B viruses.
Collapse
|
43
|
Kim KH, Li Z, Bhatnagar N, Subbiah J, Park BR, Shin CH, Pushko P, Wang BZ, Kang SM. Universal protection against influenza viruses by multi-subtype neuraminidase and M2 ectodomain virus-like particle. PLoS Pathog 2022; 18:e1010755. [PMID: 36006890 PMCID: PMC9409530 DOI: 10.1371/journal.ppat.1010755] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022] Open
Abstract
Annual influenza vaccination is recommended to update the variable hemagglutinin antigens. Here, we first designed a virus-like particle (VLP) displaying consensus multi-neuraminidase (NA) subtypes (cN1, cN2, B cNA) and M2 ectodomain (M2e) tandem repeat (m-cNA-M2e VLP). Vaccination of mice with m-cNA-M2e VLP induced broad NA inhibition (NAI), and M2e antibodies as well as interferon-gamma secreting T cell responses. Mice vaccinated with m-cNA-M2e VLP were protected against influenza A (H1N1, H5N1, H3N2, H9N2, H7N9) and influenza B (Yamagata and Victoria lineage) viruses containing substantial antigenic variations. Protective immune contributors include cellular and humoral immunity as well as antibody-dependent cellular cytotoxicity. Furthermore, comparable cross protection by m-cNA-M2e VLP vaccination was induced in aged mice. This study supports a novel strategy of developing a universal vaccine against influenza A and B viruses potentially in both young and aged populations by inducing multi-NA subtype and M2e immunity with a single VLP entity.
Collapse
Affiliation(s)
- Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Zhuo Li
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Jeeva Subbiah
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Peter Pushko
- Medigen, Inc., Frederick, Maryland, United States of America
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| |
Collapse
|
44
|
Strohmeier S, Amanat F, Carreño JM, Krammer F. Monoclonal antibodies targeting the influenza virus N6 neuraminidase. Front Immunol 2022; 13:944907. [PMID: 35967389 PMCID: PMC9363587 DOI: 10.3389/fimmu.2022.944907] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Influenza A viruses are a diverse species that include 16 true hemagglutinin (HA) subtypes and 9 true neuraminidase (NA) subtypes. While the antigenicity of many HA subtypes is reasonably well studied, less is known about NA antigenicity, especially when it comes to non-human subtypes that only circulate in animal reservoirs. The N6 subtype NAs are mostly found in viruses infecting birds. However, they have also been identified in viruses that infect mammals, such as swine and seals. More recently, highly pathogenic H5N6 subtype viruses have caused rare infections and mortality in humans. Here, we generated murine mAbs to the N6 NA, characterized their breadth and antiviral properties in vitro and in vivo and mapped their epitopes by generating escape mutant viruses. We found that the antibodies had broad reactivity across the American and Eurasian N6 lineages, but relatively little binding to the H5N6 NA. Several of the antibodies exhibited strong NA inhibition activity and some also showed activity in the antibody dependent cellular cytotoxicity reporter assay and neutralization assay. In addition, we generated escape mutant viruses for six monoclonal antibodies and found mutations on the lateral ridge of the NA. Lastly, we observed variable protection in H4N6 mouse challenge models when the antibodies were given prophylactically.
Collapse
Affiliation(s)
- Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Fatima Amanat
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness (C-VARPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Florian Krammer,
| |
Collapse
|
45
|
Strohmeier S, Amanat F, Campbell JD, Traquina P, Coffman RL, Krammer F. A CpG 1018 adjuvanted neuraminidase vaccine provides robust protection from influenza virus challenge in mice. NPJ Vaccines 2022; 7:81. [PMID: 35869085 PMCID: PMC9305062 DOI: 10.1038/s41541-022-00486-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/11/2022] [Indexed: 11/10/2022] Open
Abstract
Influenza virus infections pose a significant threat to global health. Vaccination is the main countermeasure against influenza virus spread, however, the effectiveness of vaccines is variable. Current seasonal influenza virus vaccines mostly rely on the immunodominant hemagglutinin (HA) glycoprotein on the viral surface, which usually leads to a narrow and strain-specific immune response. The HA undergoes constant antigenic drift, which can lead to a dramatic loss in vaccine effectiveness, requiring the annual reformulation and readministration of influenza virus vaccines. Recently, it has been demonstrated that the subdominant glycoprotein, neuraminidase (NA), is an attractive target for vaccine development. Here, we tested a newly developed recombinant influenza virus N1 neuraminidase vaccine candidate, named N1-MPP, adjuvanted with CpG 1018, a TLR9 agonist. Additionally, N2-MPP and B-NA-MPP vaccine constructs have been generated to cover the range of influenza viruses that are seasonally circulating in humans. These constructs have been characterized in vitro and in vivo regarding their functionality and protective potential. Furthermore, a trivalent NA-MPP mix was tested. No antigenic competition between the individual NA constructs was detected. By adjuvating the recombinant protein constructs with CpG 1018 it was possible to induce a strong and robust immune response against the NA, which provided full protection against morbidity and mortality after high lethal challenges in vivo. This study provides important insights for the development of a broadly protective NA-based influenza virus vaccine candidate.
Collapse
Affiliation(s)
- Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
46
|
Gao J, Li X, Klenow L, Malik T, Wan H, Ye Z, Daniels R. Antigenic comparison of the neuraminidases from recent influenza A vaccine viruses and 2019-2020 circulating strains. NPJ Vaccines 2022; 7:79. [PMID: 35835790 PMCID: PMC9283437 DOI: 10.1038/s41541-022-00500-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Although viral-based influenza vaccines contain neuraminidase (NA or N) antigens from the recommended seasonal strains, NA is not extensively evaluated like hemagglutinin (H) during the strain selection process. Here, we compared the antigenicity of NAs from recently recommended H1N1 (2010–2021 seasons) and H3N2 (2015–2021 seasons) vaccine strains and viruses that circulated between September 2019 and December 2020. The antigenicity was evaluated by measuring NA ferret antisera titers that provide 50% inhibition of NA activity in an enzyme-linked lectin assay. Our results show that NAs from circulating H1N1 viruses and vaccine strains for the 2017–2021 seasons are all antigenically similar and distinct from the NA in the H1N1 strain recommended for the 2010–2017 seasons. Changes in N1 antigenicity were attributed to the accumulation of substitutions over time, especially the loss of an N-linked glycosylation site (Asn386) in current N1s. The NAs from circulating H3N2 viruses and the 2020–2021 vaccine strains showed similar antigenicity that varied across the N2s in the 2016–2020 vaccine strains and was distinct from the N2 in the 2015–2016 vaccine strain. These data suggest that the recent N1 antigenicity has remained similar since the loss of the head domain N-linked glycosylation site, whereas N2 antigenicity has changed more incrementally each season.
Collapse
Affiliation(s)
- Jin Gao
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Xing Li
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Tahir Malik
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Hongquan Wan
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Zhiping Ye
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Robert Daniels
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA.
| |
Collapse
|
47
|
Tsybalova LM, Stepanova LA, Ramsay ES, Vasin AV. Influenza B: Prospects for the Development of Cross-Protective Vaccines. Viruses 2022; 14:1323. [PMID: 35746794 PMCID: PMC9228933 DOI: 10.3390/v14061323] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 01/04/2023] Open
Abstract
In this review, we analyze the epidemiological and ecological features of influenza B, one of the most common and severe respiratory infections. The review presents various strategies for cross-protective influenza B vaccine development, including recombinant viruses, virus-like particles, and recombinant proteins. We provide an overview of viral proteins as cross-protective vaccine targets, along with other updated broadly protective vaccine strategies. The importance of developing such vaccines lies not only in influenza B prevention, but also in the very attractive prospect of eradicating the influenza B virus in the human population.
Collapse
Affiliation(s)
- Liudmila M. Tsybalova
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
| | - Liudmila A. Stepanova
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
| | - Edward S. Ramsay
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
| | - Andrey V. Vasin
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
- Research Institute of Influenza named after A.A. Smorodintsev, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251 St. Petersburg, Russia
| |
Collapse
|
48
|
Chen TH, Chen CC, Wu SC. Neuraminidase (NA) 370-Loop Mutations of the 2009 Pandemic H1N1 Viruses Affect NA Enzyme Activity, Hemagglutination Titer, Mouse Virulence, and Inactivated-Virus Immunogenicity. Viruses 2022; 14:v14061304. [PMID: 35746775 PMCID: PMC9230709 DOI: 10.3390/v14061304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/23/2022] Open
Abstract
Hemagglutinin (HA) and neuraminidase (NA) are the two major envelope proteins of influenza viruses. The spatial organization of HA and NA on the virus surface needs to be optimized to promote viral fitness, host specificity, transmissibility, infectivity, and virulence. We previously demonstrated that the recombinant NA protein of the 2009 pandemic H1N1 (pH1N1) with the I365T/S366N mutation in the NA 370-loop elicited higher NA-inhibition antibody titers against the homologous pH1N1 virus and three heterologous H5N1, H3N2, and H7N9 viruses in mice. In this study, we used PR8-based reverse genetics (RG) by replacing the HA and NA genes of A/Texas/05/2009 pH1N1 virus to obtain the wild-type pH1N1 and three NA 370-loop mutant viruses of pH1N1 (I365T/S366N), RG pH1N1 (I365E/S366D), and RG pH1N1 (I365T/S366A). Our results revealed that the viral NA enzyme activity increased for the RG pH1N1(I365T/S366N) and RG pH1N1 (I365E/S366D) viruses but reduced for the RG pH1N1 (I365T/S366A) virus. The increased or decreased NA enzyme activity was found to correlate with the increase or decrease in HA titers of these NA 370-loop mutant viruses. All of these three NA 370-loop mutant RG pH1N1 viruses were less virulent than the wild-type RG pH1N1 virus in mice. Immunizations with the inactivated viruses carrying the three NA 370-loop mutations and the wild-type RG pH1N1 virus were found to elicit approximately the same titers of NA-inhibition antibodies against H1N1 and H5N1 viruses. These results may provide information for developing NA-based influenza virus vaccines.
Collapse
Affiliation(s)
- Ting-Hsuan Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Chung-Chu Chen
- Department of Internal Medicine, MacKay Memorial Hospital, Hsinchu 30071, Taiwan;
- Teaching Center of Natural Science, Minghsin University of Science and Technology, Hsinchu 30401, Taiwan
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan;
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Adimmune Corporation, Taichung 42723, Taiwan
- Correspondence:
| |
Collapse
|
49
|
Carascal MB, Pavon RDN, Rivera WL. Recent Progress in Recombinant Influenza Vaccine Development Toward Heterosubtypic Immune Response. Front Immunol 2022; 13:878943. [PMID: 35663997 PMCID: PMC9162156 DOI: 10.3389/fimmu.2022.878943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Flu, a viral infection caused by the influenza virus, is still a global public health concern with potential to cause seasonal epidemics and pandemics. Vaccination is considered the most effective protective strategy against the infection. However, given the high plasticity of the virus and the suboptimal immunogenicity of existing influenza vaccines, scientists are moving toward the development of universal vaccines. An important property of universal vaccines is their ability to induce heterosubtypic immunity, i.e., a wide immune response coverage toward different influenza subtypes. With the increasing number of studies and mounting evidence on the safety and efficacy of recombinant influenza vaccines (RIVs), they have been proposed as promising platforms for the development of universal vaccines. This review highlights the current progress and advances in the development of RIVs in the context of heterosubtypic immunity induction toward universal vaccine production. In particular, this review discussed existing knowledge on influenza and vaccine development, current hemagglutinin-based RIVs in the market and in the pipeline, other potential vaccine targets for RIVs (neuraminidase, matrix 1 and 2, nucleoprotein, polymerase acidic, and basic 1 and 2 antigens), and deantigenization process. This review also provided discussion points and future perspectives in looking at RIVs as potential universal vaccine candidates for influenza.
Collapse
Affiliation(s)
- Mark B Carascal
- Pathogen-Host-Environment Interactions Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines.,Clinical and Translational Research Institute, The Medical City, Pasig City, Philippines
| | - Rance Derrick N Pavon
- Pathogen-Host-Environment Interactions Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Windell L Rivera
- Pathogen-Host-Environment Interactions Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| |
Collapse
|
50
|
Handabile C, Sekiya T, Nomura N, Ohno M, Kawakita T, Shingai M, Kida H. Inactivated Whole Virus Particle Influenza Vaccine Induces Anti-Neuraminidase Antibodies That May Contribute to Cross-Protection against Heterologous Virus Infection. Vaccines (Basel) 2022; 10:804. [PMID: 35632561 PMCID: PMC9147865 DOI: 10.3390/vaccines10050804] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022] Open
Abstract
Despite the use of vaccines, seasonal influenza remains a risk to public health. We previously proposed the inactivated whole virus particle vaccine (WPV) as an alternative to the widely used split vaccine (SV) for the control of seasonal and pandemic influenza based on the superior priming potency of WPV to that of SV. In this study, we further examined and compared the immunological potency of monovalent WPV and SV of A/California/7/2009 (X-179A) (H1N1) pdm09 (CA/09) to generate immune responses against heterologous viruses, A/Singapore/GP1908/2015 (IVR-180) (H1N1) pdm09 (SG/15), and A/duck/Hokkaido/Vac-3/2007 (H5N1) (DH/07) in mice. Following challenge with a lethal dose of heterologous SG/15, lower virus titer in the lungs and milder weight loss were observed in WPV-vaccinated mice than in SV-vaccinated ones. To investigate the factors responsible for the differences in the protective effect against SG/15, the sera of vaccinated mice were analyzed by hemagglutination-inhibition (HI) and neuraminidase-inhibition (NI) assays to evaluate the antibodies induced against viral hemagglutinin (HA) and neuraminidase (NA), respectively. While the two vaccines induced similar levels of HI antibodies against SG/15 after the second vaccination, only WPV-vaccinated mice induced significantly higher titers of NI antibodies against the strain. Furthermore, given the significant elevation of NI antibody titers against DH/07, an H5N1 avian influenza virus, WPV was also demonstrated to induce NA-inhibiting antibodies that recognize NA of divergent strains. This could be explained by the higher conservation of epitopes of NA among strains than for HA. Taking these findings together, NA-specific antibodies induced by WPV may have contributed to better protection from infection with heterologous influenza virus SG/15, compared with SV. The present results indicate that WPV is an effective vaccine for inducing antibodies against both HA and NA of heterologous viruses and may be a useful vaccine to conquer vaccine strain mismatch.
Collapse
Affiliation(s)
- Chimuka Handabile
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (C.H.); (T.S.); (N.N.); (M.O.); (T.K.); (M.S.)
| | - Toshiki Sekiya
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (C.H.); (T.S.); (N.N.); (M.O.); (T.K.); (M.S.)
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0020, Japan
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Naoki Nomura
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (C.H.); (T.S.); (N.N.); (M.O.); (T.K.); (M.S.)
| | - Marumi Ohno
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (C.H.); (T.S.); (N.N.); (M.O.); (T.K.); (M.S.)
| | - Tomomi Kawakita
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (C.H.); (T.S.); (N.N.); (M.O.); (T.K.); (M.S.)
| | - Masashi Shingai
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (C.H.); (T.S.); (N.N.); (M.O.); (T.K.); (M.S.)
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0020, Japan
| | - Hiroshi Kida
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (C.H.); (T.S.); (N.N.); (M.O.); (T.K.); (M.S.)
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0020, Japan
- Collaborating Research Center for the Control of Infectious Diseases, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|