1
|
Wiggins KB, Winston SM, Reeves IL, Gaevert J, Spence Y, Brimble MA, Livingston B, Morton CL, Thomas PG, Sant AJ, Ross TM, Davidoff AM, Schultz-Cherry S. rAAV expressing a COBRA-designed influenza hemagglutinin generates a protective and durable adaptive immune response with a single dose. J Virol 2024; 98:e0078124. [PMID: 39078191 PMCID: PMC11338075 DOI: 10.1128/jvi.00781-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 07/31/2024] Open
Abstract
Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines. IMPORTANCE Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.
Collapse
Affiliation(s)
- Kristin B. Wiggins
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stephen M. Winston
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Isaiah L. Reeves
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Jessica Gaevert
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Yunyu Spence
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Mark A. Brimble
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Brandi Livingston
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Christopher L. Morton
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Paul G. Thomas
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Andrea J. Sant
- David H. Smith Center
for Vaccine Biology and Immunology, Department of Microbiology and
Immunology, University of Rochester Medical
Center, Rochester, New
York, USA
| | - Ted M. Ross
- Department of
Infectious Biology, Cleveland Clinic,
Cleveland, Ohio, USA
- Cleveland Clinic,
Florida Research and Innovation Center,
Port St. Lucie, Florida,
USA
| | - Andrew M. Davidoff
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of Surgery,
St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| | - Stacey Schultz-Cherry
- St. Jude Graduate
School of Biomedical Sciences,
Memphis, Tennessee, USA
- Department of
Host-Microbe Interactions, St. Jude Children’s Research
Hospital, Memphis,
Tennessee, USA
| |
Collapse
|
2
|
Zhang X, Shi H, Hendy DA, Bachelder EM, Ainslie KM, Ross TM. Multi-COBRA hemagglutinin formulated with cGAMP microparticles elicits protective immune responses against influenza viruses. mSphere 2024; 9:e0016024. [PMID: 38920382 PMCID: PMC11288037 DOI: 10.1128/msphere.00160-24] [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: 02/26/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
In humans, seasonal influenza viruses cause epidemics. Avian influenza viruses are of particular concern because they can infect multiple species and lead to unpredictable and severe disease. Therefore, there is an urgent need for a universal influenza vaccine that provides protection against all influenza strains. The cyclic GMP-AMP (cGAMP) is a promising adjuvant for subunit vaccines, which promotes type I interferons' production through the stimulator of interferon genes (STING) pathway. The encapsulation of cGAMP in acetalated dextran (Ace-DEX) microparticles (MPs) enhances its intracellular delivery. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to generate H1, H3, and H5 vaccine candidates. Monovalent and multivalent COBRA HA vaccines formulated with cGAMP Ace-DEX MPs were evaluated in mice for protective antibody responses. cGAMP MPs adjuvanted COBRA HA vaccines elicited robust antigen-specific antibodies following vaccination. Compared with COBRA HA vaccine groups with no adjuvant or blank MPs, the cGAMP MPs enhanced HAI activity elicited by COBRA HA vaccines. The HAI activity was not significantly different between cGAMP MPs adjuvanted monovalent or multivalent COBRA HA vaccines. The cGAMP MPs adjuvanted COBRA vaccine groups had higher antigen-specific IgG2a-binding titers than the COBRA vaccine groups with no adjuvant or blank MPs. The COBRA vaccines formulated with cGAMP MPs mitigated diseases caused by influenza viral challenge and decreased pulmonary viral titers in mice. Therefore, the formulation of COBRA vaccines plus cGAMP MPs is a promising universal influenza vaccine that elicits protective immune responses against human seasonal and pre-pandemic strains. IMPORTANCE Influenza viruses cause severe respiratory disease, particularly in the very young and the elderly. Next-generation influenza vaccines are needed to protect against new influenza variants. This report used a promising adjuvant, cyclic GMP-AMP (cGAMP), to enhance the elicited antibodies by an improved influenza hemagglutinin candidate and protect against influenza virus infection. Overall, adding adjuvants to influenza vaccines is an effective method to improve vaccines.
Collapse
Affiliation(s)
- Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, Florida, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
3
|
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
|
4
|
Allen JD, Ross TM. mRNA vaccines encoding computationally optimized hemagglutinin elicit protective antibodies against future antigenically drifted H1N1 and H3N2 influenza viruses isolated between 2018-2020. Front Immunol 2024; 15:1334670. [PMID: 38533508 PMCID: PMC10963417 DOI: 10.3389/fimmu.2024.1334670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/12/2024] [Indexed: 03/28/2024] Open
Abstract
Background The implementation of mRNA vaccines against COVID-19 has successfully validated the safety and efficacy of the platform, while at the same time revealing the potential for their applications against other infectious diseases. Traditional seasonal influenza vaccines often induce strain specific antibody responses that offer limited protection against antigenically drifted viruses, leading to reduced vaccine efficacy. Modern advances in viral surveillance and sequencing have led to the development of in-silico methodologies for generating computationally optimized broadly reactive antigens (COBRAs) to improve seasonal influenza vaccines. Methods In this study, immunologically naïve mice were intramuscularly vaccinated with mRNA encoding H1 and H3 COBRA hemagglutinins (HA) or wild-type (WT) influenza HAs encapsulated in lipid nanoparticles (LNPs). Results Mice vaccinated with H1 and H3 COBRA HA-encoding mRNA vaccines generated robust neutralizing serum antibody responses against more antigenically distinct contemporary and future drifted H1N1 and H3N2 influenza strains than those vaccinated with WT H1 and H3 HA-encoding mRNA vaccines. The H1 and H3 COBRA HA-encoding mRNA vaccines also prevented influenza illness, including severe disease in the mouse model against H1N1 and H3N2 viruses. Conclusions This study highlights the potential benefits of combining universal influenza antigen design technology with modern vaccine delivery platforms and exhibits how these vaccines can be advantageous over traditional WT vaccine antigens at eliciting superior protective antibody responses against a broader number of influenza virus isolates.
Collapse
Affiliation(s)
- James D Allen
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, United States
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, United States
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
5
|
Zhang X, Shi H, Hendy DA, Bachelder EM, Ainslie KM, Ross TM. Multi-COBRA hemagglutinin formulated with cGAMP microparticles elicit protective immune responses against influenza viruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582355. [PMID: 38464191 PMCID: PMC10925245 DOI: 10.1101/2024.02.27.582355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Influenza viruses cause a common respiratory disease known as influenza. In humans, seasonal influenza viruses can lead to epidemics, with avian influenza viruses of particular concern because they can infect multiple species and lead to unpredictable and severe disease. Therefore, there is an urgent need for a universal influenza vaccine that provides protection against seasonal and pre-pandemic influenza virus strains. The cyclic GMP-AMP (cGAMP) is a promising adjuvant for subunit vaccines that promotes type I interferons production through the stimulator of interferon genes (STING) pathway. The encapsulation of cGAMP in acetalated dextran (Ace-DEX) microparticles (MPs) enhances its intracellular delivery. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to generate H1, H3, and H5 vaccine candidates. Monovalent and multivalent COBRA HA vaccines formulated with cGAMP Ace-DEX MPs were evaluated in a mouse model for antibody responses and protection against viral challenge. Serological analysis showed that cGAMP MPs adjuvanted monovalent and multivalent COBRA vaccines elicited robust antigen-specific antibody responses after a prime-boost vaccination and antibody titers were further enhanced after second boost. Compared to COBRA vaccine groups with no adjuvant or blank MPs, the cGAMP MPs enhanced HAI antibody responses against COBRA vaccination. The HAI antibody titers were not significantly different between cGAMP MPs adjuvanted monovalent and multivalent COBRA vaccine groups for most of the viruses tested in panels. The cGAMP MPs adjuvanted COBRA vaccines groups had higher antigen-specific IgG2a binding titers than the COBRA vaccine groups with no adjuvant or blank MPs. The COBRA vaccines formulated with cGAMP MPs mitigated disease caused by influenza viral challenge and decreased pulmonary viral titers in mice. Therefore, the formulation of COBRA vaccines plus cGAMP MPs is a promising universal influenza vaccine that elicits protective immune responses against human seasonal and pre-pandemic strains.
Collapse
Affiliation(s)
- Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
- Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, 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
|
6
|
Uno N, Ross TM. Multivalent next generation influenza virus vaccines protect against seasonal and pre-pandemic viruses. Sci Rep 2024; 14:1440. [PMID: 38228649 PMCID: PMC10792005 DOI: 10.1038/s41598-023-51024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 12/29/2023] [Indexed: 01/18/2024] Open
Abstract
Each year, new influenza virus vaccine formulations are generated to keep up with continuously circulating and mutating viral variants. A next-generation influenza virus vaccine would provide long-lasting, broadly-reactive immune protection against current and future influenza virus strains for both seasonal and pre-pandemic viruses. Next generation immunogens were designed using computationally optimized broadly reactive antigen (COBRA) methodology to protect against a broad range of strains over numerous seasons. Novel HA and NA amino acid sequences were derived from multilayered consensus sequence alignment for multiple subtypes of influenza. This multivalent formulation was hypothesized to elicit broadly protective immune responses against both seasonal and pre-pandemic influenza viruses. Mice were vaccinated with multivalent mixtures of HA and NA (H1, H2, H3, H5, H7, N1, N2) proteins. Multivalent COBRA vaccinations elicited antibodies that recognized a broad panel of strains and vaccinated mice were protected against viruses representing multiple subtypes. This is a promising candidate for a universal influenza vaccine that elicits protective immune responses against seasonal and pre-pandemic strains over multiple seasons.
Collapse
Affiliation(s)
- Naoko Uno
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 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 Florida, 9801 SW Discovery Way, Port Saint Lucie, FL, 34986, USA.
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
7
|
Carlock MA, Ross TM. A computationally optimized broadly reactive hemagglutinin vaccine elicits neutralizing antibodies against influenza B viruses from both lineages. Sci Rep 2023; 13:15911. [PMID: 37741893 PMCID: PMC10517972 DOI: 10.1038/s41598-023-43003-2] [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: 07/04/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Influenza B viruses (IBV) can cause severe disease and death much like influenza A viruses (IAV), with a disproportionate number of infections in children. Despite moving to a quadrivalent vaccine to include strains from both the B/Victoria and B/Yamagata lineages, vaccine effectiveness rates continue to be variable and low in many past seasons. To develop more effective influenza B virus vaccines, three novel IBV hemagglutinin (HA) vaccines were designed using a computationally optimized broadly reactive antigen (COBRA) methodology. These IBV HA proteins were expressed on the surface of a virus-like particle (VLP) and used to vaccinate ferrets that were pre-immune to historical B/Victoria or B/Yamagata lineage viruses. Ferrets vaccinated with B-COBRA HA vaccines had neutralizing antibodies with high titer HAI titer against all influenza B viruses regardless of pre-immunization history. Conversely, VLPs expressing wild-type IBV HA antigens preferentially boosted titers against viruses from the same lineage and there was little-to-no seroprotective antibodies detected in ferrets with mismatched IBV pre-immune infections. Overall, a single IBV HA developed using the COBRA methodology elicited protective broadly-reactive antibodies against current and future drifted IBVs from both lineages.
Collapse
Affiliation(s)
- Michael A Carlock
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Global Vaccine Development, Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, 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.
- Global Vaccine Development, Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA.
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Global Vaccine Development, Florida Research and Innovation Center, Cleveland Clinic, 9801 SW Discovery Way, Port Saint Lucie, FL, 34987, USA.
| |
Collapse
|
8
|
Zhang H, Li Z, Zhang H, Guo Y, Zhang X, Zhang L, Yang L, Li S, Li C, Cui D, Xie R, Li Y, Huang J. Recombinant hemagglutinin displaying on yeast reshapes congenital lymphocyte subsets to prompt optimized systemic immune protection against avian influenza infection. Front Microbiol 2023; 14:1153922. [PMID: 37323887 PMCID: PMC10264594 DOI: 10.3389/fmicb.2023.1153922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/25/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Prophylactic vaccination is regarded as the most effective means to control avian flu infection. Currently, there is a need for a universal vaccine that provides broad and long-lasting protection against influenza virus. Meanwhile, although yeast-based vaccines have been used in clinic, studies are still required to further understand the molecular mechanism of yeast-based vaccines under physiological conditions. Methods We generated a yeast-based vaccine against influenza hemagglutinin (HA) of H5, H7 and H9 using surface displaying technology and evaluated the protective efficacy of chickens after exposure to H9N2 influenza virus. Results Oral yeast vaccine provided less clinical syndrome, reduced viral loading and alleviated airway damage significantly. Compared to the commercial inactivated vaccine, yeast vaccine stimulated the activation of splenic NK and APCs cells and boosted TLR7-IRF7-IFN signaling in spleen. Meanwhile, γδ T cells in the bursa of Fabricius were activated and the innate lymphoid cells (ILCs) in the bursa of Fabricius promoted the CILPs to differentiate to ILC3 cells in oral yeast birds. Moreover, the reshaped gut microbiota and a suppressed Th17-IL17-mediated inflammation in intestine was observed in oral yeast chickens, which might facilitate the recovery of intestinal mucosal immunity upon virus infection. Collectively, our findings suggest that oral yeast based multivalent bird flu vaccines provide an attractive strategy to update host defense function via reshapes of multi-systemic immune homeostasis.
Collapse
Affiliation(s)
- Han Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Zexing Li
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Huixia Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Yanyu Guo
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Xinyi Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Lilin Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Liu Yang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Shujun Li
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Changyan Li
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Daqing Cui
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Ruyu Xie
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Yongqing Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, China
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, China
| |
Collapse
|
9
|
Badten A, Ramirez A, Hernandez-Davies JE, Albin TJ, Jain A, Nakajima R, Felgner J, Davies DH, Wang SW. Protein Nanoparticle-Mediated Delivery of Recombinant Influenza Hemagglutinin Enhances Immunogenicity and Breadth of the Antibody Response. ACS Infect Dis 2023; 9:239-252. [PMID: 36607269 PMCID: PMC9926493 DOI: 10.1021/acsinfecdis.2c00362] [Citation(s) in RCA: 2] [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: 07/08/2022] [Indexed: 01/07/2023]
Abstract
The vast majority of seasonal influenza vaccines administered each year are derived from virus propagated in eggs using technology that has changed little since the 1930s. The immunogenicity, durability, and breadth of response would likely benefit from a recombinant nanoparticle-based approach. Although the E2 protein nanoparticle (NP) platform has been previously shown to promote effective cell-mediated responses to peptide epitopes, it has not yet been reported to deliver whole protein antigens. In this study, we synthesized a novel maleimido tris-nitrilotriacetic acid (NTA) linker to couple protein hemagglutinin (HA) from H1N1 influenza virus to the E2 NP, and we evaluated the HA-specific antibody responses using protein microarrays. We found that recombinant H1 protein alone is immunogenic in mice but requires two boosts for IgG to be detected and is strongly IgG1 (Th2) polarized. When conjugated to E2 NPs, IgG2c is produced leading to a more balanced Th1/Th2 response. Inclusion of the Toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) significantly enhances the immunogenicity of H1-E2 NPs while retaining the Th1/Th2 balance. Interestingly, broader homo- and heterosubtypic cross-reactivity is also observed for conjugated H1-E2 with MPLA, compared to unconjugated H1 with or without MPLA. These results highlight the potential of an NP-based delivery of HA for tuning the immunogenicity, breadth, and Th1/Th2 balance generated by recombinant HA-based vaccination. Furthermore, the modularity of this protein-protein conjugation strategy may have utility for future vaccine development against other human pathogens.
Collapse
Affiliation(s)
- Alexander
J. Badten
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Aaron Ramirez
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Jenny E. Hernandez-Davies
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Tyler J. Albin
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Aarti Jain
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Rie Nakajima
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Jiin Felgner
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - D. Huw Davies
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| | - Szu-Wen Wang
- Department
of Chemical and Biomolecular Engineering, Vaccine Research and Development
Center, Department of Physiology and Biophysics, Department of Chemistry, Department of Biomedical
Engineering, Chao Family Comprehensive Cancer Center, Institute for Immunology, University of California, Irvine, California 92697, United States
| |
Collapse
|
10
|
Chu KB, Quan FS. Respiratory Viruses and Virus-like Particle Vaccine Development: How Far Have We Advanced? Viruses 2023; 15:v15020392. [PMID: 36851606 PMCID: PMC9965150 DOI: 10.3390/v15020392] [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: 12/12/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
With technological advancements enabling globalization, the intercontinental transmission of pathogens has become much easier. Respiratory viruses are one such group of pathogens that require constant monitoring since their outbreak leads to massive public health crises, as exemplified by the influenza virus, respiratory syncytial virus (RSV), and the recent coronavirus disease 2019 (COVID-19) outbreak caused by the SARS-CoV-2. To prevent the transmission of these highly contagious viruses, developing prophylactic tools, such as vaccines, is of considerable interest to the scientific community. Virus-like particles (VLPs) are highly sought after as vaccine platforms for their safety and immunogenicity profiles. Although several VLP-based vaccines against hepatitis B and human papillomavirus have been approved for clinical use by the United States Food and Drug Administration, VLP vaccines against the three aforementioned respiratory viruses are lacking. Here, we summarize the most recent progress in pre-clinical and clinical VLP vaccine development. We also outline various strategies that contributed to improving the efficacy of vaccines against each virus and briefly discuss the stability aspect of VLPs that makes it a highly desired vaccine platform.
Collapse
Affiliation(s)
- Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence:
| |
Collapse
|
11
|
Ge P, Ross TM. Evaluation of Pre-Pandemic Trivalent COBRA HA Vaccine in Mice Pre-Immune to Historical H1N1 and H3N2 Influenza Viruses. Viruses 2023; 15:203. [PMID: 36680243 PMCID: PMC9861495 DOI: 10.3390/v15010203] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Initial exposure to influenza virus(es) during early childhood produces protective antibodies that may be recalled following future exposure to subsequent viral infections or vaccinations. Most influenza vaccine research studies use immunologically naïve animal models to assess vaccine effectiveness. However, most people have an extensive influenza immune history, with memory cells produced by viruses or vaccines representing multiple influenza viruses. In this study, we explored the effect influenza seasonal virus-induced immunity has on pre-pandemic influenza virus vaccination. The mice that were pre-immune to historical H1N1 and H3N2 seasonal influenza viruses were vaccinated with adjuvanted pre-pandemic (H2, H5, and H7) HA-based computationally optimized broadly reactive antigen (COBRA) vaccines, and were fully protected from lethal challenge, whereas the mock-vaccinated mice, with or without pre-immunity, were not protected from morbidity or mortality. Detectable antibody titers were present in the pre-immune mice vaccinated with a single dose of vaccine, but not in the immunologically naïve mice. The mice vaccinated twice with the trivalent COBRA HA vaccine had similar antibody titers regardless of their pre-immune status. Overall, seasonal pre-immunity did not interfere with the immune responses elicited by pre-pandemic COBRA HA vaccines or the protection against pre-pandemic viruses.
Collapse
Affiliation(s)
- Pan Ge
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
12
|
Chiba S, Hatta M, Pattinson D, Yasuhara A, Neumann G, Kawaoka Y. Ferret model to mimic the sequential exposure of humans to historical H3N2 influenza viruses. Vaccine 2023; 41:590-597. [PMID: 36517323 DOI: 10.1016/j.vaccine.2022.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Mutations accumulate in influenza A virus proteins, especially in the main epitopes on the virus surface glycoprotein hemagglutinin (HA). For influenza A(H3N2) viruses, in particular, the antigenicity of their HA has altered since their emergence in 1968, requiring changes of vaccine strains every few years. Most adults have been exposed to several antigenically divergent H3N2 viruses through infection and/or vaccination, and those exposures affect the immune responses of those individuals. However, animal models reflecting this 'immune history' in humans are lacking and naïve animals are generally used for vaccination and virus challenge studies. Here, we describe a ferret model to mimic the serial exposure of humans to antigenically different historical H3HA proteins. In this model, ferrets were sequentially immunized with adjuvanted recombinant H3HA proteins from two or three different H3HA antigenic clusters in chronological order, and serum neutralizing antibody titers were examined against the homologous virus and viruses from different antigenic clusters. For ferrets immunized with a single HA antigen, serum neutralizing antibody titers were elevated specifically against the homologous virus. However, after immunization with the second or third antigenically distinct HA antigen in chronological order, the ferrets showed an increase in more broadly cross-reactive neutralizing titers against the antigenically distinct viruses and against the homologous virus. Sequentially immunized animals challenged with an antigenically advanced H3N2 virus showed attenuated virus growth and less body temperature increase compared with naïve animals. These results suggest that sequential exposure to antigenically different HAs elicits broader neutralizing activity in sera and enhances immune responses against more antigenically distinct viruses Our findings may partly explain why adults who have been exposed to antigenically divergent HAs are less likely to be infected with influenza virus and have severe symptoms than children.
Collapse
Affiliation(s)
- Shiho Chiba
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI 53711, USA
| | - Masato Hatta
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI 53711, USA
| | - David Pattinson
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI 53711, USA
| | - Atsuhiro Yasuhara
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Gabriele Neumann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI 53711, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, WI 53711, USA; Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan; The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan.
| |
Collapse
|
13
|
Ontiveros-Padilla L, Batty CJ, Hendy DA, Pena ES, Roque JA, Stiepel RT, Carlock MA, Simpson SR, Ross TM, Abraham SN, Staats HF, Bachelder EM, Ainslie KM. Development of a broadly active influenza intranasal vaccine adjuvanted with self-assembled particles composed of mastoparan-7 and CpG. Front Immunol 2023; 14:1103765. [PMID: 37033992 PMCID: PMC10081679 DOI: 10.3389/fimmu.2023.1103765] [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: 11/20/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Currently licensed vaccine adjuvants offer limited mucosal immunity, which is needed to better combat respiratory infections such as influenza. Mast cells (MCs) are emerging as a target for a new class of mucosal vaccine adjuvants. Here, we developed and characterized a nanoparticulate adjuvant composed of an MC activator [mastoparan-7 (M7)] and a TLR ligand (CpG). This novel nanoparticle (NP) adjuvant was co-formulated with a computationally optimized broadly reactive antigen (COBRA) for hemagglutinin (HA), which is broadly reactive against influenza strains. M7 was combined at different ratios with CpG and tested for in vitro immune responses and cytotoxicity. We observed significantly higher cytokine production in dendritic cells and MCs with the lowest cytotoxicity at a charge-neutralizing ratio of nitrogen/phosphate = 1 for M7 and CpG. This combination formed spherical NPs approximately 200 nm in diameter with self-assembling capacity. Mice were vaccinated intranasally with COBRA HA and M7-CpG NPs in a prime-boost-boost schedule. Vaccinated mice had significantly higher antigen-specific antibody responses (IgG and IgA) in serum and mucosa compared with controls. Splenocytes from vaccinated mice had significantly increased cytokine production upon antigen recall and the presence of central and effector memory T cells in draining lymph nodes. Finally, co-immunization with NPs and COBRA HA induced influenza H3N2-specific HA inhibition antibody titers across multiple strains and partially protected mice from a challenge against an H3N2 virus. These results illustrate that the M7-CpG NP adjuvant combination can induce a protective immune response with a broadly reactive influenza antigen via mucosal vaccination.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Erik S. Pena
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
| | - John A. Roque
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rebeca T. Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael A. Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Sean R. Simpson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ted M. Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Soman N. Abraham
- Departments of Pathology, Molecular Genetics and Microbiology and Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Herman F. Staats
- Department of Pathology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccines Institute, School of Medicine, Duke University, Durham, NC, United States
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Kristy M. Ainslie,
| |
Collapse
|
14
|
Abbadi N, Nagashima K, Pena-Briseno A, Ross TM, Mousa JJ. Differential Recognition of Computationally Optimized H3 Hemagglutinin Influenza Vaccine Candidates by Human Antibodies. J Virol 2022; 96:e0089622. [PMID: 35916534 PMCID: PMC9400492 DOI: 10.1128/jvi.00896-22] [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: 06/08/2022] [Accepted: 07/10/2022] [Indexed: 02/04/2023] Open
Abstract
Among circulating influenza viruses in humans, H3N2 viruses typically evolve faster than other subtypes and have caused disease in millions of people since emerging in 1968. Computationally optimized broadly reactive antigen (COBRA) technology is one strategy to broaden vaccine-elicited antibody responses among influenza subtypes. In this study, we determined the structural integrity of an H3N2 COBRA hemagglutinin (HA), TJ5, and we probed the antigenic profile of several H3N2 COBRA HAs by assessing recognition of these immunogens by human B cells from seasonally vaccinated human subjects. Of three recently described COBRA H3 HA antigens (TJ5, NG2, and J4), we determined that TJ5 and J4 HA proteins recognize pre-existing B cells more effectively than NG2 HA and a wild-type Hong Kong/4801/2014 protein. We also isolated a panel of 12 H3 HA-specific human monoclonal antibodies (MAbs) and identified that most MAbs recognize both wild-type and COBRA HA proteins and have functional activity against a broad panel of H3N2 viruses. Most MAbs target the receptor-binding site, and one MAb targets the HA stem. MAb TJ5-5 recognizes TJ5 and J4 COBRA HA proteins but has poor recognition of NG2 HA, similar to the global B-cell analysis. We determined a 3.4 Å structure via cryo-electron microscopy of Fab TJ5-5 complexed with the H3 COBRA TJ5, which revealed residues important to the differential binding. Overall, these studies determined that COBRA H3 HA proteins have correct antigenic and structural features, and the proteins are recognized by B cells and MAbs isolated from seasonally vaccinated humans. IMPORTANCE Vaccine development for circulating influenza viruses, particularly for the H3N2 subtype, remains challenging due to consistent antigenic drift. Computationally optimized broadly reactive antigen (COBRA) technology has proven effective for broadening influenza hemagglutinin (HA)-elicited antibody responses compared to wild-type immunogens. Here, we determined the structural features and antigenic profiles of H3 COBRA HA proteins. Two H3 COBRA HA proteins, TJ5 and J4, are better recognized by pre-existing B cells and monoclonal antibodies from the 2017 to 2018 vaccine season compared to COBRA NG2 and a wild-type A/Hong Kong/2014 HA protein. We determined a cryo-electron microscopy (cryo-EM) structure of one MAb that poorly recognizes NG2, MAb TJ5-5, in complex with the TJ5 COBRA HA protein and identified residues critical to MAb recognition. As NG2 is more effective than TJ5 for the recent Hong Kong/2019 virus, these data provide insights into the diminished effectiveness of influenza vaccines across vaccine seasons.
Collapse
Affiliation(s)
- Nada Abbadi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Kaito Nagashima
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Alma Pena-Briseno
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Ted M. Ross
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jarrod J. Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
15
|
AboulFotouh K, Uno N, Xu H, Moon C, Sahakijpijarn S, Christensen DJ, Davenport GJ, Cano C, Ross TM, Williams Iii RO, Cui Z. Formulation of dry powders of vaccines containing MF59 or AddaVax by Thin-Film Freeze-Drying: Towards a dry powder universal flu vaccine. Int J Pharm 2022; 624:122021. [PMID: 35842082 DOI: 10.1016/j.ijpharm.2022.122021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 10/17/2022]
Abstract
MF59® is an oil-in-water (O/W) nanoemulsion-based vaccine adjuvant that is often used in seasonal and pandemic influenza vaccines. We explored the feasibility of developing dry powders of vaccines adjuvanted with MF59 or AddaVax™, a preclinical grade equivalent of MF59 with the same composition and droplet size as MF59, by thin-film freeze-drying (TFFD). Liquid AddaVax alone was successfully converted to a dry powder by TFFD using trehalose as a stabilizing agent while maintaining the droplet size distribution of AddaVax after it was reconstituted. TFFD was then applied to convert liquid AddaVax-adjuvanted vaccines containing either a model antigen (e.g., ovalbumin) or mono-, bi-, and tri-valent recombinant hemagglutinin (rHA) protein-based H1 and/or H3 (universal) influenza vaccine candidates, as well as the MF59-containing Fluad® Quadrivalent influenza vaccine to dry powders. Both antigens and stabilizing agents affected the physical properties of the vaccines (e.g., mean particle size and particle size distribution) after the vaccines were subjected to TFFD. Importantly, the integrity and hemagglutination activity of the rHA antigens did not significantly change and the immunogenicity of reconstituted influenza vaccine candidates was maintained when evaluated in a mouse model. The vaccine dry powder was not sensitive to repeated freezing-and-thawing, in contrast to its liquid counterpart. It is concluded that TFFD can be applied to convert liquid vaccines containing MF59 or AddaVax to dry powders while maintaining the immunogenicity of the vaccines. Ultimately, TFFD technology may be used to prepare dry powders of multivalent universal influenza vaccines.
Collapse
Affiliation(s)
- Khaled AboulFotouh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Naoko Uno
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - Haiyue Xu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Chaeho Moon
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Sawittree Sahakijpijarn
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | - Chris Cano
- TFF Pharmaceuticals, Inc., Fort Worth, TX 76107, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA; Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| | - Robert O Williams Iii
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Zhengrong Cui
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
16
|
Bivalent H1 and H3 COBRA Recombinant Hemagglutinin Vaccines Elicit Seroprotective Antibodies against H1N1 and H3N2 Influenza Viruses from 2009 to 2019. J Virol 2022; 96:e0165221. [PMID: 35289635 DOI: 10.1128/jvi.01652-21] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Commercial influenza virus vaccines often elicit strain-specific immune responses and have difficulties preventing illness caused by antigenically drifted viral variants. In the last 20 years, the H3N2 component of the annual vaccine has been updated nearly twice as often as the H1N1 component, and in 2019, a mismatch between the wild-type (WT) H3N2 vaccine strain and circulating H3N2 influenza strains led to a vaccine efficacy of ∼9%. Modern methods of developing computationally optimized broadly reactive antigens (COBRAs) for H3N2 influenza viruses utilize current viral surveillance information to design more broadly reactive vaccine antigens. Here, 7 new recombinant hemagglutinin (rHA) H3 COBRA hemagglutinin (HA) antigens were evaluated in mice. Subsequently, two candidates, J4 and NG2, were selected for further testing in influenza-preimmune animals based on their ability to elicit broadly reactive antibodies against antigenically drifted H3N2 viral isolates. In the preimmune model, monovalent formulations of J4 and NG2 elicited broadly reactive antibodies against recently circulating H3N2 influenza viruses from 2019. Bivalent mixtures of COBRA H1 and H3 rHA, Y2 + J4, and Y2 + NG2 outperformed multiple WT H1+H3 bivalent rHA mixtures by eliciting seroprotective antibodies against H1N1 and H3N2 isolates from 2009 to 2019. Overall, the newly generated COBRA HA antigens, namely, Y2, J4, and NG2, had the ability to induce broadly reactive antibodies in influenza-naive and preimmune animals in both monovalent and bivalent formulations, and these antigens outperformed H1 and H3 WT rHA vaccine antigens by eliciting seroprotective antibodies against panels of antigenically drifted historical H1N1 and H3N2 vaccine strains from 2009 to 2019. IMPORTANCE Standard-of-care influenza virus vaccines are composed of a mixture of antigens from different influenza viral subtypes. For the first time, lead COBRA H1 and H3 HA antigens, formulated as a bivalent vaccine, have been investigated in animals with preexisting immunity to influenza viruses. The cocktail of COBRA HA antigens elicited more broadly reactive anti-HA antibodies than those elicited by a comparator bivalent wild-type HA vaccine against H1 and H3 influenza viruses isolated between 2009 and 2019.
Collapse
|
17
|
Caradonna TM, Schmidt AG. Protein engineering strategies for rational immunogen design. NPJ Vaccines 2021; 6:154. [PMID: 34921149 PMCID: PMC8683408 DOI: 10.1038/s41541-021-00417-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Antibody immunodominance refers to the preferential and asymmetric elicitation of antibodies against specific epitopes on a complex protein antigen. Traditional vaccination approaches for rapidly evolving pathogens have had limited success in part because of this phenomenon, as elicited antibodies preferentially target highly variable regions of antigens, and thus do not confer long lasting protection. While antibodies targeting functionally conserved epitopes have the potential to be broadly protective, they often make up a minority of the overall repertoire. Here, we discuss recent protein engineering strategies used to favorably alter patterns of immunodominance, and selectively focus antibody responses toward broadly protective epitopes in the pursuit of next-generation vaccines for rapidly evolving pathogens.
Collapse
Affiliation(s)
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
18
|
Kotey EN, Ampofo WK, Daines R, Sadeyen JR, Iqbal M, Quaye O. Immune Response in Mice Immunized with Chimeric H1 Antigens. Vaccines (Basel) 2021; 9:1182. [PMID: 34696290 PMCID: PMC8538909 DOI: 10.3390/vaccines9101182] [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: 07/12/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022] Open
Abstract
Identification of a universal influenza vaccine candidate has remained a global challenge for both humans and animals. This study describes an approach that uses consensus sequence building to generate chimeric HAs (cHAs): two resultant H1 HA-based chimeras comprising of conserved sequences (within several areas spanning the head and stalk regions) of H1 and H5 or H9 HAs. These cHAs expressed in Drosophila cells (S2) were used to immunize mice. All immunized mice were protected from an infectious H1 virus challenge. Seroconverted mice sera to the H1 cHAs inhibited both the challenge virus and an H5 virus isolate by haemagglutination inhibition (HI) assay. These findings further emphasize that cHAs induce cross-reactive antibodies against conserved areas of both head and stalk regions of the seasonal influenza A (H1N1) pdm09 virus' HA and holds potential for further development of a universal influenza vaccine.
Collapse
Affiliation(s)
- Erasmus Nikoi Kotey
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra 23321, Ghana;
- Department of Biochemistry, Cell & Molecular Biology, University of Ghana, Legon, Accra 23321, Ghana
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra 23321, Ghana;
| | - William Kwabena Ampofo
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra 23321, Ghana;
| | - Rebecca Daines
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (R.D.); (J.-R.S.); (M.I.)
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, UK
| | - Jean-Remy Sadeyen
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (R.D.); (J.-R.S.); (M.I.)
| | - Munir Iqbal
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (R.D.); (J.-R.S.); (M.I.)
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra 23321, Ghana;
- Department of Biochemistry, Cell & Molecular Biology, University of Ghana, Legon, Accra 23321, Ghana
| |
Collapse
|
19
|
Allen JD, Ross TM. Evaluation of Next-Generation H3 Influenza Vaccines in Ferrets Pre-Immune to Historical H3N2 Viruses. Front Immunol 2021; 12:707339. [PMID: 34475872 PMCID: PMC8406686 DOI: 10.3389/fimmu.2021.707339] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/21/2021] [Indexed: 11/13/2022] Open
Abstract
Each person has a unique immune history to past influenza virus infections. Exposure to influenza viruses early in life establishes memory B cell populations that influence future immune responses to influenza vaccination. Current influenza vaccines elicit antibodies that are typically strain specific and do not offer broad protection against antigenically drifted influenza strains in all age groups of people. This is particularly true for vaccine antigens of the A(H3N2) influenza virus subtype, where continual antigenic drift necessitates frequent vaccine reformulation. Broadly-reactive influenza virus vaccine antigens offer a solution to combat antigenic drift, but they also need to be equally effective in all populations, regardless of prior influenza virus exposure history. This study examined the role that pre-existing immunity plays on influenza virus vaccination. Ferrets were infected with historical A(H3N2) influenza viruses isolated from either the 1970’s, 1980’s, or 1990’s and then vaccinated with computationally optimized broadly reactive antigens (COBRA) or wild-type (WT) influenza virus like particles (VLPs) expressing hemagglutinin (HA) vaccine antigens to examine the expansion of immune breadth. Vaccines with the H3 COBRA HA antigens had more cross-reactive antibodies following a single vaccination in all three pre-immune regimens than vaccines with WT H3 HA antigens against historical, contemporary, and future drifted A(H3N2) influenza viruses. The H3 COBRA HA vaccines also induced antibodies capable of neutralizing live virus infections against modern drifted A(H3N2) strains at higher titers than the WT H3 HA vaccine comparators.
Collapse
Affiliation(s)
- James D Allen
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States.,Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
20
|
Huang Y, França MS, Allen JD, Shi H, Ross TM. Next Generation of Computationally Optimized Broadly Reactive HA Vaccines Elicited Cross-Reactive Immune Responses and Provided Protection against H1N1 Virus Infection. Vaccines (Basel) 2021; 9:793. [PMID: 34358209 PMCID: PMC8310220 DOI: 10.3390/vaccines9070793] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022] Open
Abstract
Vaccination is the best way to prevent influenza virus infections, but the diversity of antigenically distinct isolates is a persistent challenge for vaccine development. In order to conquer the antigenic variability and improve influenza virus vaccine efficacy, our research group has developed computationally optimized broadly reactive antigens (COBRAs) in the form of recombinant hemagglutinins (rHAs) to elicit broader immune responses. However, previous COBRA H1N1 vaccines do not elicit immune responses that neutralize H1N1 virus strains in circulation during the recent years. In order to update our COBRA vaccine, two new candidate COBRA HA vaccines, Y2 and Y4, were generated using a new seasonal-based COBRA methodology derived from H1N1 isolates that circulated during 2013-2019. In this study, the effectiveness of COBRA Y2 and Y4 vaccines were evaluated in mice, and the elicited immune responses were compared to those generated by historical H1 COBRA HA and wild-type H1N1 HA vaccines. Mice vaccinated with the next generation COBRA HA vaccines effectively protected against morbidity and mortality after infection with H1N1 influenza viruses. The antibodies elicited by the COBRA HA vaccines were highly cross-reactive with influenza A (H1N1) pdm09-like viruses isolated from 2009 to 2021, especially with the most recent circulating viruses from 2019 to 2021. Furthermore, viral loads in lungs of mice vaccinated with Y2 and Y4 were dramatically reduced to low or undetectable levels, resulting in minimal lung injury compared to wild-type HA vaccines following H1N1 influenza virus infection.
Collapse
Affiliation(s)
- Ying Huang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (Y.H.); (J.D.A.); (H.S.)
| | - Monique S. França
- Poultry Diagnostic and Research Center, Department of Population Health, University of Georgia, Athens, GA 30602, USA;
| | - James D. Allen
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (Y.H.); (J.D.A.); (H.S.)
| | - Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (Y.H.); (J.D.A.); (H.S.)
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (Y.H.); (J.D.A.); (H.S.)
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
21
|
Spruit CM, Nemanichvili N, Okamatsu M, Takematsu H, Boons GJ, de Vries RP. N-Glycolylneuraminic Acid in Animal Models for Human Influenza A Virus. Viruses 2021; 13:815. [PMID: 34062844 PMCID: PMC8147317 DOI: 10.3390/v13050815] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022] Open
Abstract
The first step in influenza virus infection is the binding of hemagglutinin to sialic acid-containing glycans present on the cell surface. Over 50 different sialic acid modifications are known, of which N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc) are the two main species. Animal models with α2,6 linked Neu5Ac in the upper respiratory tract, similar to humans, are preferred to enable and mimic infection with unadapted human influenza A viruses. Animal models that are currently most often used to study human influenza are mice and ferrets. Additionally, guinea pigs, cotton rats, Syrian hamsters, tree shrews, domestic swine, and non-human primates (macaques and marmosets) are discussed. The presence of NeuGc and the distribution of sialic acid linkages in the most commonly used models is summarized and experimentally determined. We also evaluated the role of Neu5Gc in infection using Neu5Gc binding viruses and cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH)-/- knockout mice, which lack Neu5Gc and concluded that Neu5Gc is unlikely to be a decoy receptor. This article provides a base for choosing an appropriate animal model. Although mice are one of the most favored models, they are hardly naturally susceptible to infection with human influenza viruses, possibly because they express mainly α2,3 linked sialic acids with both Neu5Ac and Neu5Gc modifications. We suggest using ferrets, which resemble humans closely in the sialic acid content, both in the linkages and the lack of Neu5Gc, lung organization, susceptibility, and disease pathogenesis.
Collapse
Affiliation(s)
- Cindy M. Spruit
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.M.S.); (G.-J.B.)
| | - Nikoloz Nemanichvili
- Division of Pathology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands;
| | - Masatoshi Okamatsu
- Laboratory of Microbiology, Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Hokkaido, Japan;
| | - Hiromu Takematsu
- Department of Molecular Cell Biology, Faculty of Medical Technology, Graduate School of Health Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake 470-1192, Aichi, Japan;
| | - Geert-Jan Boons
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.M.S.); (G.-J.B.)
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Robert P. de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (C.M.S.); (G.-J.B.)
| |
Collapse
|
22
|
Strategies Targeting Hemagglutinin as a Universal Influenza Vaccine. Vaccines (Basel) 2021; 9:vaccines9030257. [PMID: 33805749 PMCID: PMC7998911 DOI: 10.3390/vaccines9030257] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/17/2022] Open
Abstract
Influenza virus has significant viral diversity, both through antigenic drift and shift, which makes development of a vaccine challenging. Current influenza vaccines are updated yearly to include strains predicted to circulate in the upcoming influenza season, however this can lead to a mismatch which reduces vaccine efficacy. Several strategies targeting the most abundant and immunogenic surface protein of influenza, the hemagglutinin (HA) protein, have been explored. These strategies include stalk-directed, consensus-based, and computationally derived HA immunogens. In this review, we explore vaccine strategies which utilize novel antigen design of the HA protein to improve cross-reactive immunity for development of a universal influenza vaccine.
Collapse
|
23
|
Broadly Reactive H2 Hemagglutinin Vaccines Elicit Cross-Reactive Antibodies in Ferrets Preimmune to Seasonal Influenza A Viruses. mSphere 2021; 6:6/2/e00052-21. [PMID: 33692193 PMCID: PMC8546680 DOI: 10.1128/msphere.00052-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Influenza vaccines have traditionally been tested in naive mice and ferrets. However, humans are first exposed to influenza viruses within the first few years of their lives. Therefore, there is a pressing need to test influenza virus vaccines in animal models that have been previously exposed to influenza viruses before being vaccinated. In this study, previously described H2 computationally optimized broadly reactive antigen (COBRA) hemagglutinin (HA) vaccines (Z1 and Z5) were tested in influenza virus “preimmune” ferret models. Ferrets were infected with historical, seasonal influenza viruses to establish preimmunity. These preimmune ferrets were then vaccinated with either COBRA H2 HA recombinant proteins or wild-type H2 HA recombinant proteins in a prime-boost regimen. A set of naive preimmune or nonpreimmune ferrets were also vaccinated to control for the effects of the multiple different preimmunities. All of the ferrets were then challenged with a swine H2N3 influenza virus. Ferrets with preexisting immune responses influenced recombinant H2 HA-elicited antibodies following vaccination, as measured by hemagglutination inhibition (HAI) and classical neutralization assays. Having both H3N2 and H1N1 immunological memory regardless of the order of exposure significantly decreased viral nasal wash titers and completely protected all ferrets from both morbidity and mortality, including the mock-vaccinated ferrets in the group. While the vast majority of the preimmune ferrets were protected from both morbidity and mortality across all of the different preimmunities, the Z1 COBRA HA-vaccinated ferrets had significantly higher antibody titers and recognized the highest number of H2 influenza viruses in a classical neutralization assay compared to the other H2 HA vaccines. IMPORTANCE H1N1 and H3N2 influenza viruses have cocirculated in the human population since 1977. Nearly every human alive today has antibodies and memory B and T cells against these two subtypes of influenza viruses. H2N2 influenza viruses caused the 1957 global pandemic and people born after 1968 have never been exposed to H2 influenza viruses. It is quite likely that a future H2 influenza virus could transmit within the human population and start a new global pandemic, since the majority of people alive today are immunologically naive to viruses of this subtype. Therefore, an effective vaccine for H2 influenza viruses should be tested in an animal model with previous exposure to influenza viruses that have circulated in humans. Ferrets were infected with historical influenza A viruses to more accurately mimic the immune responses in people who have preexisting immune responses to seasonal influenza viruses. In this study, preimmune ferrets were vaccinated with wild-type (WT) and COBRA H2 recombinant HA proteins in order to examine the effects that preexisting immunity to seasonal human influenza viruses have on the elicitation of broadly cross-reactive antibodies from heterologous vaccination.
Collapse
|
24
|
Next generation methodology for updating HA vaccines against emerging human seasonal influenza A(H3N2) viruses. Sci Rep 2021; 11:4554. [PMID: 33654128 PMCID: PMC7925519 DOI: 10.1038/s41598-020-79590-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/10/2020] [Indexed: 01/31/2023] Open
Abstract
While vaccines remain the best tool for preventing influenza virus infections, they have demonstrated low to moderate effectiveness in recent years. Seasonal influenza vaccines typically consist of wild-type influenza A and B viruses that are limited in their ability to elicit protective immune responses against co-circulating influenza virus variant strains. Improved influenza virus vaccines need to elicit protective immune responses against multiple influenza virus drift variants within each season. Broadly reactive vaccine candidates potentially provide a solution to this problem, but their efficacy may begin to wane as influenza viruses naturally mutate through processes that mediates drift. Thus, it is necessary to develop a method that commercial vaccine manufacturers can use to update broadly reactive vaccine antigens to better protect against future and currently circulating viral variants. Building upon the COBRA technology, nine next-generation H3N2 influenza hemagglutinin (HA) vaccines were designed using a next generation algorithm and design methodology. These next-generation broadly reactive COBRA H3 HA vaccines were superior to wild-type HA vaccines at eliciting antibodies with high HAI activity against a panel of historical and co-circulating H3N2 influenza viruses isolated over the last 15 years, as well as the ability to neutralize future emerging H3N2 isolates.
Collapse
|
25
|
Le Sage V, Jones JE, Kormuth KA, Fitzsimmons WJ, Nturibi E, Padovani GH, Arevalo CP, French AJ, Avery AJ, Manivanh R, McGrady EE, Bhagwat AR, Lauring AS, Hensley SE, Lakdawala SS. Pre-existing heterosubtypic immunity provides a barrier to airborne transmission of influenza viruses. PLoS Pathog 2021; 17:e1009273. [PMID: 33600489 PMCID: PMC7891786 DOI: 10.1371/journal.ppat.1009273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/04/2021] [Indexed: 12/04/2022] Open
Abstract
Human-to-human transmission of influenza viruses is a serious public health threat, yet the precise role of immunity from previous infections on the susceptibility to airborne infection is still unknown. Using the ferret model, we examined the roles of exposure duration and heterosubtypic immunity on influenza transmission. We demonstrate that a 48 hour exposure is sufficient for efficient transmission of H1N1 and H3N2 viruses. To test pre-existing immunity, a gap of 8–12 weeks between primary and secondary infections was imposed to reduce innate responses and ensure robust infection of donor animals with heterosubtypic viruses. We found that pre-existing H3N2 immunity did not significantly block transmission of the 2009 H1N1pandemic (H1N1pdm09) virus to immune animals. Surprisingly, airborne transmission of seasonal H3N2 influenza strains was abrogated in recipient animals with H1N1pdm09 pre-existing immunity. This protection from natural infection with H3N2 virus was independent of neutralizing antibodies. Pre-existing immunity with influenza B virus did not block H3N2 virus transmission, indicating that the protection was likely driven by the adaptive immune response. We demonstrate that pre-existing immunity can impact susceptibility to heterologous influenza virus strains, and implicate a novel correlate of protection that can limit the spread of respiratory pathogens through the air. Influenza viruses pose a major public health threat through both seasonal epidemics and sporadic pandemics. An individual’s first influenza virus infection leaves long-lasting immunity, which plays an unknown role on susceptibility to airborne transmission of new viral strains. We show that pre-existing heterosubtypic immunity against the 2009 H1N1 pandemic virus protects recipient animals from airborne transmission of a seasonal H3N2 influenza virus, which is independent of cross-neutralizing antibodies. Pre-existing immunity with influenza B viruses was not protective suggesting that this phenomenon is driven by an adaptive response. Taken together, these data indicate that pre-existing immunity is an important barrier to airborne transmission and can influence the emergence and spread of potentially pandemic viruses.
Collapse
Affiliation(s)
- Valerie Le Sage
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jennifer E. Jones
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Karen A. Kormuth
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - William J. Fitzsimmons
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eric Nturibi
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Gabriella H. Padovani
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Claudia P. Arevalo
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Andrea J. French
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Annika J. Avery
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Richard Manivanh
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Elizabeth E. McGrady
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Amar R. Bhagwat
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Adam S. Lauring
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Seema S. Lakdawala
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
26
|
Computationally Optimized Broadly Reactive H2 HA Influenza Vaccines Elicited Broadly Cross-Reactive Antibodies and Protected Mice from Viral Challenges. J Virol 2020; 95:JVI.01526-20. [PMID: 33115871 DOI: 10.1128/jvi.01526-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 10/25/2020] [Indexed: 01/10/2023] Open
Abstract
Influenza viruses have caused numerous pandemics throughout human history. The 1957 influenza pandemic was initiated by an H2N2 influenza virus. This H2N2 influenza virus was the result of a reassortment event between a circulating H2N2 avian virus and the seasonal H1N1 viruses in humans. Previously, our group has demonstrated the effectiveness of hemagglutinin (HA) antigens derived using computationally optimized broadly reactive antigen (COBRA) methodology against H1N1, H3N2, and H5N1 viruses. Using the COBRA methodology, H2 HA COBRA antigens were designed using sequences from H2N2 viruses isolated from humans in the 1950s and 1960s, as well as H2Nx viruses isolated from avian and mammalian species between the 1950s and 2016. In this study, the effectiveness of H2 COBRA HA antigens (Z1, Z3, Z5, and Z7) was evaluated in DBA/2J mice and compared to that of wild-type H2 HA antigens. The COBRA HA vaccines elicited neutralizing antibodies to the majority of viruses in our H2 HA panel and across all three clades as measured by hemagglutination inhibition (HAI) and neutralization assays. Comparatively, several wild-type HA vaccines elicited antibodies against a majority of the viruses in the H2 HA panel. DBA/2J mice vaccinated with COBRA vaccines showed increase survival for all three viral challenges compared to the wild-type H2 vaccines. In particular, the Z1 COBRA is a promising candidate for future work toward a pandemic H2 influenza vaccine.IMPORTANCE H2N2 influenza has caused at least one pandemic in the past. Given that individuals born after 1968 have not been exposed to H2N2 influenza viruses, a future pandemic caused by H2 influenza is likely. An effective H2 influenza vaccine would need to elicit broadly cross-reactive antibodies to multiple H2 influenza viruses. Choosing a wild-type virus to create a vaccine may elicit a narrow immune response and not protect against multiple H2 influenza viruses. COBRA H2 HA vaccines were developed and evaluated in mice along with wild-type H2 HA vaccines. Multiple COBRA H2 HA vaccines protected mice from all three viral challenges and produced broadly cross-reactive neutralizing antibodies to H2 influenza viruses.
Collapse
|
27
|
Chepkwony S, Parys A, Vandoorn E, Chiers K, Van Reeth K. Efficacy of Heterologous Prime-Boost Vaccination with H3N2 Influenza Viruses in Pre-Immune Individuals: Studies in the Pig Model. Viruses 2020; 12:v12090968. [PMID: 32882956 PMCID: PMC7552030 DOI: 10.3390/v12090968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/25/2022] Open
Abstract
In a previous study in influenza-naïve pigs, heterologous prime-boost vaccination with monovalent, adjuvanted whole inactivated vaccines (WIV) based on the European swine influenza A virus (SwIAV) strain, A/swine/Gent/172/2008 (G08), followed by the US SwIAV strain, A/swine/Pennsylvania/A01076777/2010 (PA10), was shown to induce broadly cross-reactive hemagglutination inhibition (HI) antibodies against 12 out of 15 antigenically distinct H3N2 influenza strains. Here, we used the pig model to examine the efficacy of that particular heterologous prime-boost vaccination regimen, in individuals with pre-existing infection-immunity. Pigs were first inoculated intranasally with the human H3N2 strain, A/Nanchang/933/1995. Seven weeks later, they were vaccinated intramuscularly with G08 followed by PA10 or vice versa. We examined serum antibody responses against the hemagglutinin and neuraminidase, and antibody-secreting cell (ASC) responses in peripheral blood, draining lymph nodes, and nasal mucosa (NMC), in ELISPOT assays. Vaccination induced up to 10-fold higher HI antibody titers than in naïve pigs, with broader cross-reactivity, and protection against challenge with an antigenically distant H3N2 strain. It also boosted ASC responses in lymph nodes and NMC. Our results show that intramuscular administration of WIV can lead to enhanced antibody responses and cross-reactivity in pre-immune subjects, and recall of ASC responses in lymph nodes and NMC.
Collapse
Affiliation(s)
- Sharon Chepkwony
- Laboratory of Virology, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Ghent University, 9820 Merelbeke, Belgium; (S.C.); (A.P.); (E.V.)
| | - Anna Parys
- Laboratory of Virology, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Ghent University, 9820 Merelbeke, Belgium; (S.C.); (A.P.); (E.V.)
| | - Elien Vandoorn
- Laboratory of Virology, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Ghent University, 9820 Merelbeke, Belgium; (S.C.); (A.P.); (E.V.)
| | - Koen Chiers
- Laboratory of Veterinary Pathology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium;
| | - Kristien Van Reeth
- Laboratory of Virology, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Ghent University, 9820 Merelbeke, Belgium; (S.C.); (A.P.); (E.V.)
- Correspondence: ; Tel.: +32-92647369
| |
Collapse
|
28
|
Zepeda-Cervantes J, Ramírez-Jarquín JO, Vaca L. Interaction Between Virus-Like Particles (VLPs) and Pattern Recognition Receptors (PRRs) From Dendritic Cells (DCs): Toward Better Engineering of VLPs. Front Immunol 2020; 11:1100. [PMID: 32582186 PMCID: PMC7297083 DOI: 10.3389/fimmu.2020.01100] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Virus-like particles (VLPs) have been shown to be strong activators of dendritic cells (DCs). DCs are the most potent antigen presenting cells (APCs) and their activation prompts the priming of immunity mediators based on B and T cells. The first step for the activation of DCs is the binding of VLPs to pattern recognition receptors (PRRs) on the surface of DCs, followed by VLP internalization. Like wild-type viruses, VLPs use specific PRRs from the DC; however, these recognition interactions between VLPs and PRRs from DCs have not been thoroughly reviewed. In this review, we focused on the interaction between proteins that form VLPs and PRRs from DCs. Several proteins that form VLP contain glycosylations that allow the direct interaction with PRRs sensing carbohydrates, prompting DC maturation and leading to the development of strong adaptive immune responses. We also discussed how the knowledge of the molecular interaction between VLPs and PRRs from DCs can lead to the smart design of VLPs, whether based on the fusion of foreign epitopes or their chemical conjugation, as well as other modifications that have been shown to induce a stronger adaptive immune response and protection against infectious pathogens of importance in human and veterinary medicine. Finally, we address the use of VLPs as tools against cancer and allergic diseases.
Collapse
Affiliation(s)
- Jesús Zepeda-Cervantes
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Josué Orlando Ramírez-Jarquín
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis Vaca
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
29
|
Ross TM. Universal Influenza Vaccine Approaches Using Full-Length or Head-Only Hemagglutinin Proteins. J Infect Dis 2020; 219:S57-S61. [PMID: 30715379 DOI: 10.1093/infdis/jiz004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There is currently an unmet need to develop an effective broadly reactive or universal vaccine against influenza viruses capable of conferring protection against both seasonal and prepandemic strains. Influenza vaccines elicit immune responses that are protective against antigenically similar viruses within a subtype. These vaccines elicit antibodies that target the surface viral glycoproteins hemagglutinin and neuraminidase. If there is an antigenic mismatch between these proteins in the influenza vaccines and cocirculating influenza isolates, there is a decrease in the vaccine effectiveness in vaccinated persons. Various novel influenza vaccine candidates are being evaluated in animal studies and clinical human trials. This article focuses on the advantages and potential shortcomings of broadly reactive or universal vaccine candidates based on the hemagglutinin globular head and the thoughts about using this antigen as the basis for future influenza vaccine strategies.
Collapse
Affiliation(s)
- Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens.,Department of Infectious Diseases, University of Georgia, Athens
| |
Collapse
|
30
|
Skarlupka AL, Ross TM. Immune Imprinting in the Influenza Ferret Model. Vaccines (Basel) 2020; 8:vaccines8020173. [PMID: 32276530 PMCID: PMC7348859 DOI: 10.3390/vaccines8020173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/28/2022] Open
Abstract
The initial exposure to influenza virus usually occurs during childhood. This imprinting has long-lasting effects on the immune responses to subsequent infections and vaccinations. Animal models that are used to investigate influenza pathogenesis and vaccination do recapitulate the pre-immune history in the human population. The establishment of influenza pre-immune ferret models is necessary for understanding infection and transmission and for designing efficacious vaccines.
Collapse
Affiliation(s)
- Amanda L. Skarlupka
- 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
- Correspondence: ; Tel.: +1-706-542-9708
| |
Collapse
|
31
|
An Influenza Virus Hemagglutinin Computationally Optimized Broadly Reactive Antigen Elicits Antibodies Endowed with Group 1 Heterosubtypic Breadth against Swine Influenza Viruses. J Virol 2020; 94:JVI.02061-19. [PMID: 31996430 DOI: 10.1128/jvi.02061-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
32
|
Sautto GA, Kirchenbaum GA, Abreu RB, Ecker JW, Pierce SR, Kleanthous H, Ross TM. A Computationally Optimized Broadly Reactive Antigen Subtype-Specific Influenza Vaccine Strategy Elicits Unique Potent Broadly Neutralizing Antibodies against Hemagglutinin. THE JOURNAL OF IMMUNOLOGY 2019; 204:375-385. [PMID: 31811019 DOI: 10.4049/jimmunol.1900379] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022]
Abstract
Computationally optimized broadly reactive Ags (COBRA) targeting H1 elicit a broad cross-reactive and cross-neutralizing Ab response against multiple H1N1 viral strains. To assess B cell breadth, Mus musculus (BALB/c) Ab-secreting cells elicited by a candidate COBRA hemagglutinin (HA) (termed P1) were compared with Ab-secreting cells elicited by historical H1N1 vaccine strains. In addition, to evaluate the Ab response elicited by P1 HA at increased resolution, a panel of P1 HA-specific B cell hybridomas was generated following immunization of mice with COBRA P1 and the corresponding purified mAbs were characterized for Ag specificity and neutralization activity. Both head- and stem-directed mAbs were elicited by the P1 HA Ag, with some mAbs endowed with Ab-dependent cell-mediated cytotoxicity activity. P1 HA-elicited mAbs exhibited a wide breadth of HA recognition, ranging from narrowly reactive to broadly reactive mAbs. Interestingly, we identified a P1 HA-elicited mAb (1F8) exhibiting broad hemagglutination inhibition activity against both seasonal and pandemic H1N1 influenza strains. Furthermore, mAb 1F8 recognized an overlapping, but distinct, epitope compared with other narrowly hemagglutination inhibition-positive mAbs elicited by the P1 or wild-type HA Ags. Finally, P1 HA-elicited mAbs were encoded by distinct H chain variable and L chain variable gene segment rearrangements and possessed unique CDR3 sequences. Collectively, the functional characterization of P1 HA-elicited mAbs sheds further insights into the underlying mechanism(s) of expanded Ab breadth elicited by a COBRA HA-based immunogen and advances efforts toward design and implementation of a more broadly protective influenza vaccine.
Collapse
Affiliation(s)
- Giuseppe A Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Greg A Kirchenbaum
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Rodrigo B Abreu
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Jeffrey W Ecker
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Spencer R Pierce
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | | | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602; .,Department of Infectious Diseases, University of Georgia, Athens, GA 30602
| |
Collapse
|
33
|
Kelvin AA, Zambon M. Influenza imprinting in childhood and the influence on vaccine response later in life. Euro Surveill 2019; 24:1900720. [PMID: 31796156 PMCID: PMC6891942 DOI: 10.2807/1560-7917.es.2019.24.48.1900720] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Alyson A Kelvin
- Department of Pediatrics, Division of Infectious Disease, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Canadian Centre for Vaccinology, IWK Health Centre, Halifax, Canada
| | - Maria Zambon
- National Infection Service, Public Health England, London, United Kingdom
| |
Collapse
|
34
|
Abstract
Introduction: High variance is associated with influenza vaccine effectiveness (VE). Accumulating evidence suggests that preexisting influenza-specific immunity results in the variance in VE and skews overall immune response to vaccination. Nevertheless, the investigation of preexisting immunity is highly limited due to the lack of proper methodology to explore the complex individual immune history.Areas covered: Retrospective observational studies have shown that the preexisting influenza specific immunity influences on VE. To simplify a discussion, we summarized important findings from the observational studies based on the transition of the individual immune history: the first exposure to influenza virus, the first vaccination, and repetitive exposure throughout life. We also discussed the prospectus of pre-immunized animal models to investigate the interaction between preexisting immunity and vaccine efficacy.Expert opinion: A better understanding in the underlying mechanisms on preexisting immunity is critical to improve VE and to help develop novel vaccine strategies. Using animals pre-immunized with historical influenza strains is a promising approach to verify the underlying immunologic mechanism of interaction between preexisting immunity and vaccine antigen. Also, pre-immunized animal models will be better able to evaluate the efficacy of novel vaccine strategies than naïve animals.
Collapse
Affiliation(s)
- Hyesun Jang
- 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
| |
Collapse
|
35
|
Belongia EA, McLean HQ. Influenza Vaccine Effectiveness: Defining the H3N2 Problem. Clin Infect Dis 2019; 69:1817-1823. [DOI: 10.1093/cid/ciz411] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/16/2019] [Indexed: 12/22/2022] Open
Abstract
Abstract
Observational studies have consistently shown that influenza vaccine effectiveness (VE) is lower for H3N2 relative to H1N1pdm09 and type B, and this is not entirely explained by antigenic match. The triad of virus, vaccine, and host immunity provides a framework to examine contributing factors. Antigenic evolution facilitates H3N2 immune escape, and increasing glycosylation of the hemagglutinin shields antigenic sites from antibody binding. Egg passage adaptation of vaccine viruses generates mutations that alter glycosylation, impair the neutralizing antibody response, and reduce VE. Complex host immune factors may also influence H3N2 VE, including early childhood imprinting and repeated vaccination, but their role is uncertain. Of the triad of contributing factors, only changes to the vaccine are readily achievable. However, it is unclear whether current licensed non–egg-based vaccines generate superior protection against H3N2. The optimal strategy remains to be defined, but newer vaccine technology platforms offer great potential.
Collapse
Affiliation(s)
- Edward A Belongia
- Center for Clinical Epidemiology and Population Health, Marshfield Clinic Research Institute, Wisconsin
| | - Huong Q McLean
- Center for Clinical Epidemiology and Population Health, Marshfield Clinic Research Institute, Wisconsin
| |
Collapse
|
36
|
Skarlupka AL, Owino SO, Suzuki-Williams LP, Crevar CJ, Carter DM, Ross TM. Computationally optimized broadly reactive vaccine based upon swine H1N1 influenza hemagglutinin sequences protects against both swine and human isolated viruses. Hum Vaccin Immunother 2019; 15:2013-2029. [PMID: 31448974 PMCID: PMC6773400 DOI: 10.1080/21645515.2019.1653743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/27/2019] [Accepted: 08/01/2019] [Indexed: 11/17/2022] Open
Abstract
Swine H1 influenza viruses were stable within pigs for nearly 70 years until in 1998 when a classical swine virus reassorted with avian and human influenza viruses to generate the novel triple reassortant H1N1 strain that eventually led to the 2009 influenza pandemic. Previously, our group demonstrated broad protection against a panel of human H1N1 viruses using HA antigens derived by the COBRA methodology. In this report, the effectiveness of COBRA HA antigens (SW1, SW2, SW3 and SW4), which were designed using only HA sequences from swine H1N1 and H1N2 isolates, were tested in BALB/c mice. The effectiveness of these vaccines were compared to HA sequences designed using both human and swine H1 HA sequences or human only sequences. SW2 and SW4 elicited antibodies that detected the pandemic-like virus, A/California/07/2009 (CA/09), had antibodies with HAI activity against almost all the classical swine influenza viruses isolated from 1973-2015 and all of the Eurasian viruses in our panel. However, sera collected from mice vaccinated with SW2 or SW4 had HAI activity against ~25% of the human seasonal-like influenza viruses isolated from 2009-2015. In contrast, the P1 COBRA HA vaccine (derived from both swine and human HA sequences) elicited antibodies that had HAI activity against both swine and human H1 viruses and protected against CA/09 challenge, but not a human seasonal-like swine H1N2 virus challenge. However, the SW1 vaccine protected against this challenge as well as the homologous vaccine. These results support the idea that a pan-swine-human H1 influenza virus vaccine is possible.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Computers, Molecular
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H1N1 Subtype
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Swine
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
| | - Simon O. Owino
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | | | - Corey J. Crevar
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, FL, USA
| | - Donald M. Carter
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, 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
| |
Collapse
|