1
|
Simmons HC, Finney J, Kotaki R, Adachi Y, Moseman AP, Watanabe A, Song S, Robinson-McCarthy LR, Le Sage V, Kuraoka M, Moseman EA, Kelsoe G, Takahashi Y, McCarthy KR. A protective and broadly binding antibody class engages the influenza virus hemagglutinin head at its stem interface. mBio 2025:e0089225. [PMID: 40391889 DOI: 10.1128/mbio.00892-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 05/22/2025] Open
Abstract
Influenza infection and vaccination impart strain-specific immunity that protects against neither seasonal antigenic variants nor the next pandemic. However, antibodies directed to conserved sites can confer broad protection. Here, we identify and characterize a class of human antibodies that engage a previously undescribed, conserved epitope on the influenza hemagglutinin (HA) protein. Prototype antibody S8V1-157 binds at the normally occluded interface between the HA head and stem. Antibodies to this HA head-stem interface epitope are non-neutralizing in vitro but protect against lethal influenza infection in mice. These antibodies bind to most influenza A subtypes and seasonal human variants, and are present at low frequencies in the memory B cell populations of multiple human donors. Vaccines designed to elicit these antibodies might contribute to "universal" influenza immunity. IMPORTANCE Antibodies to the influenza virus hemagglutinin (HA) protein confer the strongest protection against infection. Human antibodies elicited by infection and/or vaccination fail to protect against antigenically novel animal, pandemic, or human seasonal viruses. Improved vaccines are needed. We identify a novel class of antibodies that bind most divergent HA subtypes and all seasonal human HA antigenic variants tested. These antibodies confer protection from lethal influenza challenge in animal models. The corresponding epitope on the HA head is occluded by its interaction with the stem and is inaccessible in the well-resolved prefusion state. The immunogenicity of this head-stem interface indicates that poorly understood conformations of HA presenting widely conserved surfaces are explored in biochemical, cell-based, and in vivo assays. Head-stem interface antibodies warrant further investigation as an avenue to improve influenza vaccines and therapeutics.
Collapse
Affiliation(s)
- Holly C Simmons
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Joel Finney
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Ryutaro Kotaki
- Research Center for Vaccine Development, National Institute of Infectious Diseases, Japan Institute for Health Security, Shinjuku, Tokyo, Japan
| | - Yu Adachi
- Research Center for Vaccine Development, National Institute of Infectious Diseases, Japan Institute for Health Security, Shinjuku, Tokyo, Japan
| | - Annie Park Moseman
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Akiko Watanabe
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Shengli Song
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Lindsey R Robinson-McCarthy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Valerie Le Sage
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Masayuki Kuraoka
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - E Ashley Moseman
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Garnett Kelsoe
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Yoshimasa Takahashi
- Research Center for Vaccine Development, National Institute of Infectious Diseases, Japan Institute for Health Security, Shinjuku, Tokyo, Japan
| | - Kevin R McCarthy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
2
|
Myburgh L, van Loon K, Huijbers EJM, van Beijnum JR, Russell CA, Griffioen AW. Guided design for the development of an evolution-proof influenza vaccine. Vaccine 2025; 59:127281. [PMID: 40398325 DOI: 10.1016/j.vaccine.2025.127281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/24/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Influenza remains a significant public health concern, particularly among high-risk populations, due to its capacity to cause annual epidemics and potentially trigger global pandemics. Despite the availability of countermeasures such as vaccines and antiviral treatments, their effectiveness is hindered by factors such as resistance development with manufacturing of the influenza vaccine still heavily relying on decades-old technologies. This review therefore examines the mechanisms by which influenza viruses evade host immunity and evaluates current and emerging approaches to enhance vaccine-mediated protection. Advances targeting the conserved hemagglutinin (HA) stem, incorporating multiple HA subtypes, and the use of adjuvants are discussed, alongside increased attention to neuraminidase (NA) and other viral components as immunogenic targets. Strategic epitope prediction, through glycan masking, evolutionary forecasting, and consensus sequence design, offer promising frameworks for rational vaccine design. Furthermore, delivery platforms, including recombinant protein, mRNA, and conjugate vaccines are explored for their potential to elicit broad and durable immunity. Collectively, these developments highlight a multifaceted approach towards the design of effective interventions against this persistent healthcare challenge.
Collapse
Affiliation(s)
- Lauren Myburgh
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Karlijn van Loon
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Colin A Russell
- Laboratory of Applied Evolutionary Biology, Department of Medical Microbiology, Amsterdam, UMC, the Netherlands
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Singleton KL, Post DJ, Augustine AD, Ison MG. Collaborative influenza vaccine innovation centers (CIVICs) program. Vaccine 2025; 54:127118. [PMID: 40286588 PMCID: PMC12070419 DOI: 10.1016/j.vaccine.2025.127118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Affiliation(s)
- Kentner L Singleton
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Lane, Bethesda, MD 20852, USA
| | - Diane J Post
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Lane, Bethesda, MD 20852, USA
| | - Alison D Augustine
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Lane, Bethesda, MD 20852, USA.
| | - Michael G Ison
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Lane, Bethesda, MD 20852, USA.
| |
Collapse
|
4
|
Chen PL, Richardson RA, Rovito S, Yang G, Writt HN, Ojha C, DeBeauchamp J, Crumpton JC, Woodard K, Penaflor M, Kercher L, Webby RJ, Sautto GA, Ross TM, Russell CJ. Live-attenuated pandemic H1N1 influenza vaccines expressing computationally optimized broadly reactive antigens (COBRAs) are immunogenic and protective in mice and ferrets. Vaccine 2025; 53:127090. [PMID: 40186991 DOI: 10.1016/j.vaccine.2025.127090] [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: 12/11/2024] [Revised: 03/24/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Computationally optimized broadly reactive antigens (COBRAs) induce broad and protective immune responses across multiple viral vaccine platforms. However, their suitability for incorporation into live attenuated influenza vaccines (LAIVs) remains uncertain, as antigen modifications could potentially impact LAIV generation, replication, stability, or immunogenicity. In this study, COBRA hemagglutinin (HA) and neuraminidase (NA) antigens designated Y2 and N1I, respectively, of the influenza H1N1 subtype were reverse-engineered into A/Puerto Rico/8/1934 (PR8)-based LAIVs. The impact of HA stability and NA composition on LAIV properties was evaluated in cell culture, mice, and ferrets. COBRA LAIV yields were higher in MDCK cells compared to Vero cells, and a higher HA activation pH was associated with increased LAIV growth in cell culture. The COBRA LAIVs elicited broad antibody responses against pandemic H1N1 viruses and provided robust protection in both mice and ferrets. The standard COBRA LAIV, containing unmodified HA Y2 and NA N1I, had virus inactivation pH and HA activation pH values of 5.4 and 5.6, respectively. In contrast, a modified COBRA LAIV, containing an HA2-K153E mutation and NA from the vaccine strain A/Hawaii/70/2019 (HI19), had a virus inactivation pH of 5.3 and an elevated HA activation pH of 6.0. This modified LAIV had improved growth in cell culture and greater protection from challenge virus lung titers in elderly ferrets. These studies demonstrate the successful integration of COBRA antigen engineering into a LAIV platform. Furthermore, fine-tuning HA stability and NA composition appears to be a promising strategy to enhance LAIVs containing modifications to computationally optimized antigens.
Collapse
MESH Headings
- Animals
- Ferrets
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Mice
- Neuraminidase/immunology
- Neuraminidase/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Female
- Dogs
- Antigens, Viral/immunology
- Antigens, Viral/genetics
- Madin Darby Canine Kidney Cells
- Immunogenicity, Vaccine
- Chlorocebus aethiops
- Vero Cells
- Mice, Inbred BALB C
- Humans
Collapse
Affiliation(s)
- Po-Ling Chen
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert A Richardson
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Samuel Rovito
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Guohua Yang
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Haley N Writt
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Chet Ojha
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jennifer DeBeauchamp
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeri Carol Crumpton
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Karlie Woodard
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Melissa Penaflor
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lisa Kercher
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Richard J Webby
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Giuseppe A Sautto
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Charles J Russell
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
5
|
Liu R, Yang L, Feng J, Zhang S, Wu L, Du Y, Kong D, Xu Y, Peng T. M2e/NP Dual Epitope-Displaying Nanoparticles Enhance Cross-Protection of Recombinant HA Influenza Vaccine: A Universal Boosting Strategy. Vaccines (Basel) 2025; 13:412. [PMID: 40333315 PMCID: PMC12031538 DOI: 10.3390/vaccines13040412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025] Open
Abstract
Background/Objectives: Vaccination remains the most effective means of preventing influenza virus infections. However, the continuous antigenic drift and shift of influenza viruses lead to a reduced efficacy of the existing vaccines, necessitating vaccines capable of broad protection. Methods: To address this, we developed a modular vaccine strategy pairing a clinical-stage adjuvanted recombinant hemagglutinin (HA) vaccine (SCVC101) with OMN, a heptameric nanoparticle displaying conserved influenza A virus T-cell epitopes from nucleoprotein (NP) and matrix 2 ectodomain (M2e). Results: OMN induced cross-reactive M2e-specific antibodies, binding to diverse influenza A subtypes. Critically, the co-administration of OMN with SCVC101 enhanced cellular immunity and cross-protection without diminishing HA-induced humoral responses. Conclusions: This dual-antigen delivery system enables annual HA component updates, aligned with WHO recommendations, while the conserved OMN nanoparticle acts as a universal booster, leveraging existing production infrastructure. This approach offers a promising strategy for improving the influenza vaccine's efficacy against emerging viral variants.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; (R.L.); (L.Y.); (J.F.); (S.Z.)
| | - Lejun Yang
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; (R.L.); (L.Y.); (J.F.); (S.Z.)
| | - Jin Feng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; (R.L.); (L.Y.); (J.F.); (S.Z.)
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510663, China; (L.W.); (Y.X.)
| | - Songchen Zhang
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; (R.L.); (L.Y.); (J.F.); (S.Z.)
| | - Liping Wu
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510663, China; (L.W.); (Y.X.)
| | - Yingying Du
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; (R.L.); (L.Y.); (J.F.); (S.Z.)
| | - Dexin Kong
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; (R.L.); (L.Y.); (J.F.); (S.Z.)
| | - Yuhua Xu
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510663, China; (L.W.); (Y.X.)
| | - Tao Peng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; (R.L.); (L.Y.); (J.F.); (S.Z.)
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510663, China; (L.W.); (Y.X.)
| |
Collapse
|
6
|
Yuan Y, Yu H, Agrahari AK, Gao J, Kang H, Daniels R, Chen X. Catch, Cut, or Block? Versatile 4-N-Derivatized Sialyl Glycosides for Influenza Virus Neuraminidase Detection and Purification. Angew Chem Int Ed Engl 2025:e202505903. [PMID: 40210607 DOI: 10.1002/anie.202505903] [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: 03/13/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/12/2025]
Abstract
Seasonal influenza continues to threaten human lives and pose a significant burden to healthcare systems and the economy, emphasizing the need for developing better influenza vaccines, diagnostics, and antiviral therapeutics. To address these challenges, we generated a library of structurally diverse sialyl glycosides containing 4-N-derivatized sialic acids by a highly efficient one-pot two-enzyme chemoenzymatic sialylation strategy. Sialosides containing 4-azido-substituted sialic acid were selectively cleaved by sialidases from influenza viruses, whereas sialosides containing 4-acetamido-modified sialic acid were resistant to sialidase cleavage. Interestingly, sialosides containing 4-amino- or 4-guanidino-substituted sialic acid were effective inhibitors moderately or highly resistant to cleavage by influenza sialidases (also called neuraminidases). The sialosides containing the 4-guanidino-substituted sialic acid represent a new class of sialidase substrate analog-based inhibitors. We took advantage of this unique property to create a ligand-based approach for efficiently isolating influenza virions from egg allantoic fluid with high purity. Together, these compounds are versatile probes and ligands for developing new approaches to detect, profile, isolate, and characterize influenza viruses via neuraminidases on their surface.
Collapse
Affiliation(s)
- Yue Yuan
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA
| | - Hai Yu
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA
| | - Anand Kumar Agrahari
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA
| | - Jin Gao
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hyeog Kang
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Robert Daniels
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Xi Chen
- Department of Chemistry, University of California, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
7
|
Park H, Kingstad-Bakke B, Cleven T, Jung M, Kawaoka Y, Suresh M. Diversifying T-cell responses: safeguarding against pandemic influenza with mosaic nucleoprotein. J Virol 2025; 99:e0086724. [PMID: 39898643 PMCID: PMC11915837 DOI: 10.1128/jvi.00867-24] [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: 05/17/2024] [Accepted: 12/22/2024] [Indexed: 02/04/2025] Open
Abstract
Pre-existing T-cell responses have been linked to reduced disease severity and better clinical outcomes during the 2009 influenza pandemic and the recent COVID-19 pandemic. We hypothesized that diversifying T-cell responses, particularly targeting conserved viral proteins such as the influenza A virus (IAV) nucleoprotein (NP), could protect against both epidemic and pandemic IAV strains. To test this, we created a mosaic nucleoprotein (MNP) by synthesizing a sequence that maximized the representation of 9-mer epitopes from 7422 NP sequences across human, swine, and avian IAVs. Notably, the MNP sequence showed high homology with the NP of the H5N1 strain affecting dairy cows in the ongoing outbreak. Mucosal immunization with the adjuvanted MNP vaccine induced robust CD8 and CD4 T-cell responses against both known immunodominant and in silico predicted subdominant epitopes. MNP-vaccinated mice challenged with epidemic H1N1 and H3N2 strains, which shared immunodominant CD8 and/or CD4 T-cell epitopes, showed a significant (~4 log) reduction in lung viral load. Importantly, MNP-vaccinated mice challenged with a pandemic H1N1 strain lacking shared immunodominant CD8 or CD4 epitopes exhibited a superior reduction in lung viral load, linked to T-cell responses targeting subdominant epitopes present in both the MNP and pandemic strain NP. These results suggest that a diversified T-cell response induced by the MNP vaccine could provide broad protection against severe disease from both current and emerging IAV strains. IMPORTANCE The World Health Organization (WHO) estimates that seasonal influenza causes 3-5 million cases of severe illness annually. The influenza virus frequently undergoes genetic changes through antigenic drift and antigenic shift, resulting in annual epidemics and occasional pandemics. Consequently, a major public health objective is to develop a universal influenza vaccine that offers broad protection against both current and pandemic influenza A strains. In this study, we designed a nucleoprotein (NP) antigen (termed mosaic NP) comprising antigenic regions found in thousands of influenza viruses, aiming to use it as a vaccine to induce broad anti-influenza T-cell responses. Our findings indicate that the mosaic NP vaccine provided significant protection against seasonal H1N1 and H3N2, as well as the pandemic H1N1 strain, demonstrating its effectiveness across various influenza subtypes. These findings suggest that the mosaic NP is a potential universal influenza vaccine antigen, capable of protecting against diverse strains of influenza viruses.
Collapse
Affiliation(s)
- Hongtae Park
- Department of Pathobiological Sciences, University of Wisconsin, Madison, Wisconsin, USA
| | - Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin, Madison, Wisconsin, USA
| | - Thomas Cleven
- Department of Pathobiological Sciences, University of Wisconsin, Madison, Wisconsin, USA
| | - Myunghwan Jung
- Department of Pathobiological Sciences, University of Wisconsin, Madison, Wisconsin, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin, Madison, Wisconsin, USA
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
8
|
Monto AS, Foster-Tucker JE, Callear AP, Leis AM, Godonou ET, Smith M, Truscon R, Johnson E, Thomas LJ, Thompson MS, Fry AM, Flannery B, Malosh RE, Petrie JG, Lauring AS, Martin ET. Respiratory Viral Infections From 2015 to 2022 in the HIVE Cohort of American Households: Incidence, Illness Characteristics, and Seasonality. J Infect Dis 2025; 231:795-804. [PMID: 39179953 PMCID: PMC11847950 DOI: 10.1093/infdis/jiae423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Viral respiratory illnesses are the most common acute illnesses experienced and generally follow a predicted pattern over time. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic interrupted that pattern. METHODS The HIVE (Household Influenza Vaccine Evaluation) study was established in 2010 to follow a cohort of Southeast Michigan households over time. Initially focused on influenza, surveillance was expanded to include other major respiratory pathogens, and, starting in 2015, the population was followed year round. Symptoms of acute illness were reported, and respiratory specimens were collected and tested to identify viral infections. Based on the known population being followed, virus-specific incidence was calculated. RESULTS From 2015 to 2022, 1755 participants were followed in HIVE for 7785 person-years with 7833 illnesses documented. Before the pandemic, rhinovirus (RV) and common cold human coronaviruses (HCoVs) were the viruses most frequently identified, and incidence decreased with increasing age. Type A influenza was next but with comparable incidence by age. Parainfluenza and respiratory syncytial viruses were less frequent overall, followed by human metapneumoviruses. Incidence was highest in young children, but infections were frequently documented in all age groups. Seasonality followed patterns established decades ago. The SARS-CoV-2 pandemic disrupted these patterns, except for RV and, to a lesser extent, HCoVs. In the first 2 years of the pandemic, RV incidence far exceeded that of SARS-CoV-2. CONCLUSIONS Longitudinal cohort studies are important in comparing the incidence, seasonality, and characteristics of different respiratory viral infections. Studies documented the differential effect of the pandemic on the incidence of respiratory viruses in addition to SARS-CoV-2.
Collapse
Affiliation(s)
- Arnold S Monto
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Joshua E Foster-Tucker
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Amy P Callear
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Aleda M Leis
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Elie-Tino Godonou
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Matthew Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Rachel Truscon
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Emileigh Johnson
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Lara J Thomas
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | | | - Alicia M Fry
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Brendan Flannery
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ryan E Malosh
- Division of Immunizations, Michigan Department of Health and Human Services, Lansing, Michigan, USA
| | - Joshua G Petrie
- Marshfield Clinic Research Institute, Marshfield, Wisconsin, USA
| | - Adam S Lauring
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Emily T Martin
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Deming ME, Toapanta FR, Pasetti M, Golding H, Khurana S, Hamouda T, Fattom A, Liang Y, Tennant SM, McGilvray MF, Bernal PJ, Oshinsky JJ, Datta S, Booth JP, Coughlan L, Neuzil KM, Costley CD, Kotloff KL, Sztein MB, Ortiz JR. An intranasal adjuvanted, recombinant influenza A/H5 vaccine candidate induces broad priming against diverse influenza A/H5N1 virus clades in a phase I randomized trial in healthy adults. RESEARCH SQUARE 2025:rs.3.rs-6059149. [PMID: 40092447 PMCID: PMC11908355 DOI: 10.21203/rs.3.rs-6059149/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
We conducted a randomized, controlled phase I trial (NCT05397119) of a novel adjuvanted recombinant influenza A/H5 (A/Indonesia/05/2005, clade 2.1) hemagglutinin vaccine, administered intranasally in two doses 28 days apart at three antigen levels. Control groups received unadjuvanted recombinant H5 or formulation buffer placebo. Six months later, participants received a heterologous unadjuvanted inactivated influenza A/H5N1 (A/Vietnam/1203/2004, clade 1) vaccine intramuscularly. All vaccines were safe and well tolerated. After the primary intranasal series, serum hemagglutination inhibition and microneutralization responses were minimal. Increases in mucosal and serum IgG/IgA, serum surface plasmon resonance antibody binding, memory B cell and CD4 T cell activity, and antibody-dependent cell-mediated cytotoxicity were observed only in recipients primed intranasally with adjuvanted H5 vaccine. Following the inactivated H5N1 boost, robust responses across all immune assays, as well as microneutralization responses against diverse H5N1 clades (including currently circulating clade 2.3.4.4b), occurred in adjuvanted vaccine recipients, demonstrating successful priming and broad responses.
Collapse
Affiliation(s)
- Meagan E Deming
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Marcela Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Hana Golding
- Center for Biologics Evaluation and Research (CBER), Food and Drug Administration
| | - Surender Khurana
- Center for Biologics Evaluation and Research (CBER), Food and Drug Administration
| | | | | | - Yuanyuan Liang
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Sharon M Tennant
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Megan F McGilvray
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Paula J Bernal
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Shrimati Datta
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Lynda Coughlan
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Kathleen M Neuzil
- Center for Vaccine Development, University of Maryland School of Medicine
| | | | - Karen L Kotloff
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Marcelo B Sztein
- Center for Vaccine Development, University of Maryland School of Medicine
| | - Justin R Ortiz
- Center for Vaccine Development, University of Maryland School of Medicine
| |
Collapse
|
10
|
Lane A, Quach HQ, Ovsyannikova IG, Kennedy RB, Ross TM, Einav T. Characterizing the Short- and Long-Term Temporal Dynamics of Antibody Responses to Influenza Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.26.25322965. [PMID: 40061340 PMCID: PMC11888507 DOI: 10.1101/2025.02.26.25322965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Most influenza vaccine studies evaluate acute antibody responses 1 month post-vaccination, leaving long-term immunity poorly understood. Here, we performed a combined analysis of 14 large-scale vaccine studies and conducted two new studies mapping antibody responses in high resolution from their inception out to 1 year post-vaccination. Vaccine antibody responses were classified as weak (<4x fold-change at 1 month and 1 year), transient (≥4x at 1 month, <4x at 1 year), or durable (≥4x at 1 month and 1 year). Surprisingly, >50% of vaccine recipients were weak across seasons, age groups, sexes, pre-vaccination titers, and high or standard vaccine doses. Peak fold-change at 1 month post-vaccination was strongly associated with the long-term response, with most transient responders achieving a maximum fold-change of 4x, while most durable responders reached ≥16x, with both groups maintaining these titers for 2 months (10-75 days post-vaccination). Using the weak, transient, and durable trajectories, a single time point early in the response (days 7-8 or 21) predicted an individual's response out to 1 year post-vaccination. These results demonstrate that influenza vaccine responses range from little-to-no response to eliciting strong-and-durable immunity, highlighting the stark heterogeneity that is consistently seen across influenza seasons.
Collapse
Affiliation(s)
- Aaron Lane
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Huy Q Quach
- Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Tal Einav
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
11
|
Perry SS, Brice DC, Sakr AA, Kandeil A, DeBeauchamp J, Ghonim M, Jones J, Miller L, Vegesana K, Crawford JC, Langfitt DM, Kercher L, Abdelsamed HA, Webster RG, Thomas PG, Webby RJ, Okda FA. Modulation of cytokeratin and cytokine/chemokine expression following influenza virus infection of differentiated human tonsillar epithelial cells. J Virol 2025; 99:e0146024. [PMID: 39791909 PMCID: PMC11852761 DOI: 10.1128/jvi.01460-24] [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: 08/20/2024] [Accepted: 12/08/2024] [Indexed: 01/12/2025] Open
Abstract
The tonsils have been identified as a site of replication for Epstein-Barr virus, adenovirus, human papillomavirus, and other respiratory viruses. Human tonsil epithelial cells (HTECs) are a heterogeneous group of actively differentiating cells. Here, we investigated the cellular features and susceptibility of differentiated HTECs to specific influenza viruses, including expression of avian-type and mammalian-type sialic acid (SA) receptors, viral replication dynamics, and the associated cytokine secretion profiles. We found that differentiated HTECs possess more abundant α2,3-linked SA (preferentially bound by avian influenza viruses) than α2,6-linked SA (preferentially bound by mammalian strains). This dual receptor expression suggests a role in influenza virus adaptation and tropism within the tonsils by facilitating the binding and entry of multiple influenza virus strains. Our results indicated the susceptibility of differentiated HTECs to a wide range of influenza viruses from human, swine, and avian hosts. Virus production for most strains was detected as early as 1 day post-infection (dpi), and typically peaked by 3 dpi. However, pandemic H1N1 virus showed remarkably delayed replication kinetics that did not peak until at least 7 dpi. Notably, influenza virus infection impacted the expression of cytokeratins in HTEC cultures, which correlated with altered cytokine secretion patterns. These patterns varied within the strains but were most distinct in swine H3N2 infection. In conclusion, differentiated HTECs exhibited a strain-specific pattern of influenza virus replication and innate immune responses that included changes in cytokeratin and cytokine expression. These studies shed light on the complex interplay between influenza viruses and host cells in the tonsils. IMPORTANCE To develop effective interventions against influenza, it is important to identify host factors affecting pathogenesis and immune responses. Tonsils are lymphoepithelial organs characterized by infiltration of B and T lymphocytes into the squamous epithelium of tonsillar crypts, beneath which germinal centers play key roles in antigen processing and the immune response. Influenza virus tropism in the human upper respiratory tract is a key determinant of host-range, pathogenesis, and transmission. Accordingly, experimental models using primary cells from the human respiratory tract are relevant for assessing virus tropism and replication competence. Our study addresses the dynamics of influenza virus replication in HTECs, including cellular tropism, infectivity, and cytokeratin and cytokine expression. The results of this study highlight the complex interplay between structural proteins and immune signaling pathways, all of which provide valuable insights into host-virus interactions.
Collapse
Affiliation(s)
- S. Scott Perry
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - David C. Brice
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Ahmed Atef Sakr
- Cornell Veterinary Biobank, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Ahmed Kandeil
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
- National Research Center, Giza, Egypt
| | - Jennifer DeBeauchamp
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mohamed Ghonim
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jeremy Jones
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lance Miller
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Kasi Vegesana
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jeremy Chase Crawford
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Deanna M. Langfitt
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Lisa Kercher
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hossam A. Abdelsamed
- Immunology Center of Georgia (IMMCG), Department of Physiology, Medical College of Georgia (MCG), Augusta University, Augusta, Georgia, USA
| | - Robert G. Webster
- Department of Host-Microbe Interactions, 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
| | - Richard J. Webby
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Faten A. Okda
- Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA
- National Research Center, Giza, Egypt
| |
Collapse
|
12
|
Shi H, Zhang X, Ross TM. A single dose of inactivated influenza virus vaccine expressing COBRA hemagglutinin elicits broadly-reactive and long-lasting protection. PLoS One 2025; 20:e0308680. [PMID: 39982912 PMCID: PMC11844911 DOI: 10.1371/journal.pone.0308680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/24/2024] [Indexed: 02/23/2025] Open
Abstract
Influenza virus infections present a pervasive global health concern resulting in millions of hospitalizations and thousands of fatalities annually. To address the influenza antigenic variation, the computationally optimized broadly reactive antigen (COBRA) methodology was used to design influenza hemagglutinin (HA) or neuraminidase (NA) for universal influenza vaccine candidates. In this study, whole inactivated virus (WIV) or split inactivated virus (SIV) vaccine formulations expressing either the H1 COBRA HA or H3 COBRA HA were formulated with or without an adjuvant and tested in ferrets with pre-existing anti-influenza immunity. A single dose of the COBRA-WIV vaccine elicited a robust and broadly reactive antibody response against H1N1 and H3N2 influenza viruses. In contrast, the COBRA-SIV elicited antibodies that recognized fewer viruses, but with R-DOATP, its specificity was expanded. Vaccinated ferrets were protected against morbidity and mortality following challenge with A/California/07/2009 at 14 weeks post-vaccination with reduced viral shedding post-infection compared to the naïve ferrets. However, the COBRA-IIVs did not block the viral transmission to naïve ferrets. The contact infection induced less severe disease and delayed viral shedding than direct infection. Overall, the COBRA HA WIV or the COBRA HA SIV plus R-DOTAP elicited broadly reactive antibodies with long-term protection against viral challenge and reduced viral transmission following a single dose of vaccine in ferrets pre-immune to historical H1N1 and H3N2 influenza viruses. IMPORTANCE The goal of the next-generation influenza vaccine is to provide broadly reactive protection against various drifted influenza strains. With the previous studies evaluating the COBRA HA-based vaccines, the breadth of antibody activities was confirmed following two or three vaccinations. However, for the commercial influenza vaccine, only one shot is required. In this study, only one shot was administrated to the pre-immune ferrets and the COBRA-WIV efficiently elicited broadly reactive antibodies and long-lasting protection against the pdm09 strain. Moreover, this study showed that different infection methods can lead to different disease severity, which emphasizes the significance of the model selection. The infection was conducted 14 weeks post-vaccination to evaluate the long-term protection elicited by only one vaccination. This is the first longevity study describing the immune responses elicited by COBRA-IIVs in ferrets and provides promising results for the potential clinical utilization.
Collapse
Affiliation(s)
- Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, United States of America
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States of America
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
| |
Collapse
|
13
|
Pekarek MJ, Madapong A, Wiggins J, Weaver EA. Adenoviral-Vectored Multivalent Vaccine Provides Durable Protection Against Influenza B Viruses from Victoria-like and Yamagata-like Lineages. Int J Mol Sci 2025; 26:1538. [PMID: 40004004 PMCID: PMC11855595 DOI: 10.3390/ijms26041538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Despite annual vaccines, Influenza B viruses (IBVs) continue to cause severe infections and have a significant impact and burden on the healthcare system. Improving IBV vaccine effectiveness is a key focus, with various strategies under investigation. In this research, we used a computational design to select wildtype sequences for a multivalent viral-vectored vaccine (rAd-Tri-Vic) targeting the Victoria-like (Vic) hemagglutinin (HA) protein. In mouse models, the vaccine induced strong antibody and T cell responses, providing complete protection against both lineage-specific and cross-lineage (Yamagata-like) lethal challenges. The immune responses remained robust for up to six months, which demonstrated sustained protection. These results highlight the potential of HA-targeted multivalent vaccines to enhance the IBV efficacy and address protection against antigenically diverse IBV strains.
Collapse
Affiliation(s)
- Matthew J. Pekarek
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| | - Adthakorn Madapong
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
| | - Joshua Wiggins
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| | - Eric A. Weaver
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (M.J.P.); (A.M.); (J.W.)
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| |
Collapse
|
14
|
Ekanger CT, Dinesh Kumar N, Koutstaal RW, Zhou F, Beukema M, Waldock J, Jochems SP, Mulder N, van Els CACM, Engelhardt OG, Mantel N, Buno KP, Brokstad KA, Engelsen AST, Cox RJ, Melgert BN, Huckriede ALW, van Kasteren PB. Comparison of air-liquid interface transwell and airway organoid models for human respiratory virus infection studies. Front Immunol 2025; 16:1532144. [PMID: 39981254 PMCID: PMC11839712 DOI: 10.3389/fimmu.2025.1532144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Complex in vitro respiratory models, including air-liquid interface (ALI) transwell cultures and airway organoids, have emerged as promising tools for studying human respiratory virus infections. These models address several limitations of conventional two-dimensional cell line and animal models. However, the lack of standardized protocols for the application of these models in infection studies limits the possibilities for comparing results across different research groups. Therefore, we applied a collaborative approach to harmonize several aspects of experimental methodology between different research laboratories, aiming to assess the comparability of different models of human airway epithelium in the context of respiratory viral infections. Methods In this study, we compared three different models of human respiratory epithelium: a primary human bronchial epithelial cell-derived ALI transwell model, and two airway organoid models established from human airway- and lung-derived adult stem cells. We first assessed the presence of various differentiated cell types using immunofluorescence microscopy. Using a shared stock of influenza A virus, we then assessed viral growth kinetics, epithelial cytokine responses, and serum-mediated inhibition of infection. Results The presence of club, goblet, and ciliated cells was confirmed in all models. We observed similar viral replication kinetics with a >4-log increase in virus titre across all models using a TCID50 assay. Following infection, a reproducible antiviral cytokine response, including a consistent increase in CXCL10, IL-6, IFN-λ1, IFN-λ2/3, and IFN-β, was detected across all models. Finally, neutralization was assessed by pre-incubation of virus with human serum. Reduced viral replication was observed across all models, resulting in a 3- to 6-log decrease in virus titres as quantified by TCID50. Discussion In conclusion, all three models produced consistent results regardless of the varying cell sources, culturing approaches, and infection methods. Our collaborative efforts to harmonize infection experiments and compare ALI transwell and airway organoid models described here aid in advancing our understanding and improving the standardization of these complex in vitro respiratory models for future studies.
Collapse
Affiliation(s)
- Camilla T. Ekanger
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Nilima Dinesh Kumar
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rosanne W. Koutstaal
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Martin Beukema
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joanna Waldock
- Influenza Resource Centre, Vaccines, Science Research & Innovation, Medicines and Healthcare Products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | - Simon P. Jochems
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Noa Mulder
- Groningen Research Institute for Pharmacy, Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Cécile A. C. M. van Els
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Section Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Othmar G. Engelhardt
- Influenza Resource Centre, Vaccines, Science Research & Innovation, Medicines and Healthcare Products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | | | | | - Karl Albert Brokstad
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Agnete S. T. Engelsen
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Rebecca J. Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Barbro N. Melgert
- Groningen Research Institute for Pharmacy, Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anke L. W. Huckriede
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Puck B. van Kasteren
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
15
|
Shirey KA, Joseph J, Coughlan L, Nijhuis H, Varley AW, Blanco JCG, Vogel SN. An adenoviral vector encoding an inflammation-inducible antagonist, HMGB1 Box A, as a novel therapeutic approach to inflammatory diseases. mBio 2025; 16:e0338724. [PMID: 39699172 PMCID: PMC11796352 DOI: 10.1128/mbio.03387-24] [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: 10/30/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Influenza, as well as other respiratory viruses, can trigger local and systemic inflammation resulting in an overall "cytokine storm" that produces serious outcomes such as acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). We hypothesized that gene therapy platforms could be useful in these cases if the production of an anti-inflammatory protein reflects the intensity and duration of the inflammatory condition. The recombinant protein would be produced and released only in the presence of the inciting stimulus, avoiding immunosuppression or other unwanted side effects that may occur when treating infectious diseases with anti-inflammatory drugs. To test this hypothesis, we developed AdV.C3-Tat/HIV-Box A, an inflammation-inducible cassette that remains innocuous in the absence of inflammation but releases HMGB1 Box A, an antagonist of high mobility group box 1 (HMGB1), in response to inflammatory stimuli such as lipopolysaccharide (LPS) or influenza virus infection. We report here that this novel inflammation-inducible HMGB1 Box A construct in a non-replicative adenovirus (AdV) vector mitigates lung and systemic inflammation therapeutically in response to influenza infection. We anticipate that this strategy will apply to the treatment of multiple diseases in which HMGB1-mediated signaling is a central driver of inflammation.IMPORTANCEMany inflammatory diseases are mediated by the action of a host-derived protein, HMGB1, on Toll-like receptor 4 (TLR4) to elicit an inflammatory response. We have engineered a non-replicative AdV vector that produces HMGB1 Box A, an antagonist of HMGB1-induced inflammation, under the control of an endogenous complement component C3 (C3) promoter sequence, that is inducible by LPS and influenza in vitro and ex vivo in macrophages (Mϕ) and protects mice and cotton rats therapeutically against infection with mouse-adapted and human non-adapted influenza strains, respectively, in vivo. We anticipate that this novel strategy will apply to the treatment of multiple infectious and non-infectious diseases in which HMGB1-mediated TLR4 signaling is a central driver of inflammation.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - John Joseph
- Sigmovir Biosystems Inc., Rockville, Maryland, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Haye Nijhuis
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | | | | | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Bull JJ, Koelle K, Antia R. Waning immunity drives respiratory virus evolution and reinfection. Evol Med Public Health 2025; 13:101-110. [PMID: 40443498 PMCID: PMC12121555 DOI: 10.1093/emph/eoaf002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/06/2024] [Accepted: 01/15/2025] [Indexed: 06/02/2025] Open
Abstract
Viruses differ in the number and types of host tissues in which they replicate. For example, systemically replicating viruses such as measles infect cells and tissues throughout the body, whereas respiratory viruses such as influenza viruses and coronaviruses replicate only in the respiratory tract. Reinfections with respiratory viruses are thought to be driven by ongoing antigenic immune escape in the viral population. However, this does not explain why antigenic variation is frequently observed in respiratory viruses and not systemically replicating viruses. Here, we argue that the rapid rate of waning immunity in the respiratory tract is a key driver of antigenic evolution in respiratory viruses. Waning immunity results in hosts with immunity levels that protect against homologous reinfection but are insufficient to protect against infection with an antigenically different (heterologous) strain. Thus, when partially immune hosts are present at a high enough density, an immune escape variant can invade the viral population even though that variant cannot infect solidly immune hosts. Invasion can occur even when the variant's immune escape mutation incurs a fitness cost, although any such cost is likely to be short-lived from compensatory evolution. Thus, the mutant lineage may replace the wild type and, as immunity to it builds, the process will repeat. Our model provides a new explanation for the pattern of successive emergence and replacement of antigenic variants that has been observed in many respiratory viruses. We discuss our model relative to others for understanding the drivers of antigenic evolution in respiratory viruses.
Collapse
Affiliation(s)
- James J Bull
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Katia Koelle
- Department of Biology, Emory University, Atlanta, GA, USA
- Emory Center of Excellence for Influenza Research and Response (CEIRR), Atlanta, GA, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA, USA
- Emory Center of Excellence for Influenza Research and Response (CEIRR), Atlanta, GA, USA
| |
Collapse
|
17
|
Ovchinnikov V, Karplus M. Phenomenological Modeling of Antibody Response from Vaccine Strain Composition. Antibodies (Basel) 2025; 14:6. [PMID: 39846614 PMCID: PMC11755667 DOI: 10.3390/antib14010006] [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: 11/30/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
The elicitation of broadly neutralizing antibodies (bnAbs) is a major goal of vaccine design for highly mutable pathogens, such as influenza, HIV, and coronavirus. Although many rational vaccine design strategies for eliciting bnAbs have been devised, their efficacies need to be evaluated in preclinical animal models and in clinical trials. To improve outcomes for such vaccines, it would be useful to develop methods that can predict vaccine efficacies against arbitrary pathogen variants. As a step in this direction, here, we describe a simple biologically motivated model of antibody reactivity elicited by nanoparticle-based vaccines using only antigen amino acid sequences, parametrized with a small sample of experimental antibody binding data from influenza or SARS-CoV-2 nanoparticle vaccinations. Results: The model is able to recapitulate the experimental data to within experimental uncertainty, is relatively insensitive to the choice of the parametrization/training set, and provides qualitative predictions about the antigenic epitopes exploited by the vaccine, which are testable by experiment. For the mosaic nanoparticle vaccines considered here, model results suggest indirectly that the sera obtained from vaccinated mice contain bnAbs, rather than simply different strain-specific Abs. Although the present model was motivated by nanoparticle vaccines, we also apply it to a mutlivalent mRNA flu vaccination study, and demonstrate good recapitulation of experimental results. This suggests that the model formalism is, in principle, sufficiently flexible to accommodate different vaccination strategies. Finally, we show how the model could be used to rank the efficacies of vaccines with different antigen compositions. Conclusions: Overall, this study suggests that simple models of vaccine efficacy parametrized with modest amounts of experimental data could be used to compare the effectiveness of designed vaccines.
Collapse
Affiliation(s)
- Victor Ovchinnikov
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Martin Karplus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Laboratoire de Chimie Biophysique, ISIS, Université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
18
|
Grovenstein P, Bhatnagar N, Kim KH, Pal SS, Le CTT, Raha JR, Liu R, Shin CH, Park BR, Du L, Subbiah J, Wang BZ, Kang SM. Influenza 5xM2e mRNA lipid nanoparticle vaccine confers broad immunity and significantly enhances the efficacy of inactivated split vaccination when coadministered. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:104-114. [PMID: 40073270 PMCID: PMC11844137 DOI: 10.1093/jimmun/vkae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/29/2024] [Indexed: 03/14/2025]
Abstract
Current influenza vaccines are not effective in conferring protection against antigenic variants and pandemics. To improve cross-protection of influenza vaccination, we developed a 5xM2e messenger RNA (mRNA) vaccine encoding the tandem repeat conserved ectodomain (M2e) of ion channel protein M2 derived from human, swine, and avian influenza A viruses. The lipid nanoparticle (LNP)-encapsulated 5xM2e mRNA vaccine was immunogenic, eliciting high levels of M2e-specific IgG antibodies, IFN-γ+ T cells, T follicular helper cells, germinal center phenotypic B cells, and plasma cells. The mice with 5xM2e mRNA vaccination were broadly protected against lethal infection regardless of hemagglutinin (H1, H3, H5) subtypes by preventing severe weight loss. Injection of 5xM2e mRNA LNP vaccine induced acute innate responses recruiting monocytes, macrophages, and diverse subsets of dendritic cells. A single dose of combined 5xM2e mRNA LNP and split vaccines resulted in significantly enhanced and sustainable IgG antibody responses to viral antigens and protection against homologous and heterologous viruses. This study provides a new strategy of combined mRNA and seasonal vaccination, significantly enhancing vaccine protective efficacy.
Collapse
Affiliation(s)
- Phillip Grovenstein
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Chau Thuy Tien Le
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Rong Liu
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Chong Hyun Shin
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Bo Ryoung Park
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Lanying Du
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Jeeva Subbiah
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
19
|
Peacock TP, Moncla L, Dudas G, VanInsberghe D, Sukhova K, Lloyd-Smith JO, Worobey M, Lowen AC, Nelson MI. The global H5N1 influenza panzootic in mammals. Nature 2025; 637:304-313. [PMID: 39317240 DOI: 10.1038/s41586-024-08054-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Influenza A viruses have caused more documented global pandemics in human history than any other pathogen1,2. High pathogenicity avian influenza viruses belonging to the H5N1 subtype are a leading pandemic risk. Two decades after H5N1 'bird flu' became established in poultry in Southeast Asia, its descendants have resurged3, setting off a H5N1 panzootic in wild birds that is fuelled by: (1) rapid intercontinental spread, reaching South America and Antarctica for the first time4,5; (2) fast evolution via genomic reassortment6; and (3) frequent spillover into terrestrial7,8 and marine mammals9. The virus has sustained mammal-to-mammal transmission in multiple settings, including European fur farms10,11, South American marine mammals12-15 and US dairy cattle16-19, raising questions about whether humans are next. Historically, swine are considered optimal intermediary hosts that help avian influenza viruses adapt to mammals before jumping to humans20. However, the altered ecology of H5N1 has opened the door to new evolutionary pathways. Dairy cattle, farmed mink or South American sea lions may have the potential to serve as new mammalian gateways for transmission of avian influenza viruses to humans. In this Perspective, we explore the molecular and ecological factors driving the sudden expansion in H5N1 host range and assess the likelihood of different zoonotic pathways leading to an H5N1 pandemic.
Collapse
Affiliation(s)
- Thomas P Peacock
- The Pirbright Institute, Pirbright, Woking, UK
- Department of Infectious Disease, St Mary's Medical School, Imperial College London, London, UK
| | - Louise Moncla
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gytis Dudas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - David VanInsberghe
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Center of Excellence for Influenza Research and Response (Emory-CEIRR), Atlanta, GA, USA
| | - Ksenia Sukhova
- Department of Infectious Disease, St Mary's Medical School, Imperial College London, London, UK
| | - James O Lloyd-Smith
- Department of Ecology and Evolutionary Biology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael Worobey
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
| | - Anice C Lowen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Center of Excellence for Influenza Research and Response (Emory-CEIRR), Atlanta, GA, USA
| | - Martha I Nelson
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
20
|
Shi H, Zhang X, Ge P, Meliopoulos V, Freiden P, Livingston B, Schultz-Cherry S, Ross TM. Inactivated influenza virus vaccines expressing COBRA hemagglutinin elicited broadly reactive, long-lived protective antibodies. Hum Vaccin Immunother 2024; 20:2356269. [PMID: 38826029 PMCID: PMC11152115 DOI: 10.1080/21645515.2024.2356269] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/12/2024] [Indexed: 06/04/2024] Open
Abstract
The influenza viruses cause seasonal respiratory illness that affect millions of people globally every year. Prophylactic vaccines are the recommended method to prevent the breakout of influenza epidemics. One of the current commercial influenza vaccines consists of inactivated viruses that are selected months prior to the start of a new influenza season. In many seasons, the vaccine effectiveness (VE) of these vaccines can be relatively low. Therefore, there is an urgent need to develop an improved, more universal influenza vaccine (UIV) that can provide broad protection against various drifted strains in all age groups. To meet this need, the computationally optimized broadly reactive antigen (COBRA) methodology was developed to design a hemagglutinin (HA) molecule as a new influenza vaccine. In this study, COBRA HA-based inactivated influenza viruses (IIV) expressing the COBRA HA from H1 or H3 influenza viruses were developed and characterized for the elicitation of immediate and long-term protective immunity in both immunologically naïve or influenza pre-immune animal models. These results were compared to animals vaccinated with IIV vaccines expressing wild-type H1 or H3 HA proteins (WT-IIV). The COBRA-IIV elicited long-lasting broadly reactive antibodies that had hemagglutination-inhibition (HAI) activity against drifted influenza variants.
Collapse
Affiliation(s)
- Hua Shi
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Xiaojian Zhang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Pan Ge
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
| | - Victoria Meliopoulos
- St. Jude Children’s Research Hospital, Department of Host-Microbe Interactions, Memphis, TN, USA
| | - Pam Freiden
- St. Jude Children’s Research Hospital, Department of Host-Microbe Interactions, Memphis, TN, USA
| | - Brandi Livingston
- St. Jude Children’s Research Hospital, Department of Host-Microbe Interactions, Memphis, TN, USA
| | - Stacey Schultz-Cherry
- St. Jude Children’s Research Hospital, Department of Host-Microbe Interactions, Memphis, TN, USA
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
21
|
Mallajosyula V, Chakraborty S, Sola E, Fong RF, Shankar V, Gao F, Burrell AR, Gupta N, Wagar LE, Mischel PS, Capasso R, Staat MA, Chien YH, Dekker CL, Wang TT, Davis MM. Coupling antigens from multiple subtypes of influenza can broaden antibody and T cell responses. Science 2024; 386:1389-1395. [PMID: 39700292 PMCID: PMC12036609 DOI: 10.1126/science.adi2396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/29/2024] [Accepted: 11/08/2024] [Indexed: 12/21/2024]
Abstract
The seasonal influenza vaccine contains strains of viruses from distinct subtypes that are grown independently and then combined. However, most individuals exhibit a more robust response to one of these strains and thus are vulnerable to infection by others. By studying a monozygotic twin cohort, we found that although prior exposure is a factor, host genetics are a stronger driver of subtype bias to influenza viral strains. We found that covalent coupling of heterologous hemagglutinin (HA) from different viral strains could largely eliminate subtype bias in an animal model and in a human tonsil organoid system. We proposed that coupling of heterologous antigens improves antibody responses across influenza strains by broadening T cell help, and we found that using this approach substantially improved the antibody response to avian influenza HA.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Antibodies, Viral/immunology
- Antibody Formation/immunology
- Antigens, Viral/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Influenza A virus/immunology
- Influenza A virus/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Palatine Tonsil/immunology
- Palatine Tonsil/virology
- CD4-Positive T-Lymphocytes/immunology
- Organoids/immunology
- Organoids/virology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Vamsee Mallajosyula
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Saborni Chakraborty
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Elsa Sola
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryan Furuichi Fong
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vishnu Shankar
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fei Gao
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Allison R. Burrell
- Department of Pediatrics, Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45220, USA
| | - Neha Gupta
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lisa E. Wagar
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Paul S. Mischel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Robson Capasso
- Division of Sleep Surgery, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mary A. Staat
- Department of Pediatrics, Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45220, USA
| | - Yueh-Hsiu Chien
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cornelia L. Dekker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Taia T. Wang
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Mark M. Davis
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
22
|
Taaffe J, Ostrowsky JT, Mott J, Goldin S, Friede M, Gsell P, Chadwick C. Advancing influenza vaccines: A review of next-generation candidates and their potential for global health impact. Vaccine 2024; 42:126408. [PMID: 39369576 DOI: 10.1016/j.vaccine.2024.126408] [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: 06/21/2024] [Revised: 08/20/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Influenza vaccines are an essential tool for influenza prevention, control and preparedness. However, demand for them and their programmatic suitability globally is significantly influenced by their variable effectiveness against influenza illness annually, limited duration of protection and need for yearly updating and vaccination. As such, the World Health Organization and major funders, such as the United States National Institute of Allergy and Infectious Diseases and Bill and Melinda Gates Foundation, have strongly encouraged developing influenza vaccines with increased efficacy, breadth and duration of protection. Here, we review the next-generation influenza vaccine pipeline, focusing on products in clinical development, and compare their characteristics to currently approved seasonal influenza vaccines. METHODS To identify and characterize next-generation influenza vaccine candidates, we conducted a comprehensive literature review, using the CIDRAP Universal Influenza Vaccine Technology Landscape as a primary reference source and extracting additional information from peer-reviewed manuscripts, clinical trial records and other media in the public domain. RESULTS Our analysis reveals a robust clinical development pipeline for next-generation influenza vaccines, featuring a diversity of approaches to address existing vaccine challenges and several candidates in advanced stages of development. mRNA vaccines emerged as a predominant platform, as evidenced by the number of candidates focused on improved seasonal protection as well as combination vaccine candidates targeting additional respiratory viruses. CONCLUSION While still early in development, results from universal or broadly protective products are promising and warrant continued investment from funders. As most Phase 3 candidates are mRNA-based and include combination vaccines, it is critical to begin considering how these new products may become integrated into the current global influenza vaccine strain selection and manufacturing ecosystems, and existing immunization programmes.
Collapse
Affiliation(s)
| | - Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, USA
| | - Joshua Mott
- World Health Organization, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
23
|
Khudainazarova NS, Granovskiy DL, Kondakova OA, Ryabchevskaya EM, Kovalenko AO, Evtushenko EA, Arkhipenko MV, Nikitin NA, Karpova OV. Prokaryote- and Eukaryote-Based Expression Systems: Advances in Post-Pandemic Viral Antigen Production for Vaccines. Int J Mol Sci 2024; 25:11979. [PMID: 39596049 PMCID: PMC11594041 DOI: 10.3390/ijms252211979] [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: 08/19/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
This review addresses the ongoing global challenge posed by emerging and evolving viral diseases, underscoring the need for innovative vaccine development strategies. It focuses on the modern approaches to creating vaccines based on recombinant proteins produced in different expression systems, including bacteria, yeast, plants, insects, and mammals. This review analyses the advantages, limitations, and applications of these expression systems for producing vaccine antigens, as well as strategies for designing safer, more effective, and potentially 'universal' antigens. The review discusses the development of vaccines for a range of viral diseases, excluding SARS-CoV-2, which has already been extensively studied. The authors present these findings with the aim of contributing to ongoing research and advancing the development of antiviral vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nikolai A. Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.S.K.); (D.L.G.); (O.A.K.); (E.M.R.); (A.O.K.); (E.A.E.); (M.V.A.); (O.V.K.)
| | | |
Collapse
|
24
|
Zuckermann FA, Grinkova YV, Husmann RJ, Pires-Alves M, Storms S, Chen WY, Sligar SG. An effective vaccine against influenza A virus based on the matrix protein 2 (M2). Vet Microbiol 2024; 298:110245. [PMID: 39293153 PMCID: PMC11758941 DOI: 10.1016/j.vetmic.2024.110245] [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: 03/19/2024] [Revised: 07/14/2024] [Accepted: 08/30/2024] [Indexed: 09/20/2024]
Abstract
The ever-increasing antigenic diversity of the hemagglutinin (HA) of influenza A virus (IAV) poses a significant challenge for effective vaccine development. Notably, the matrix protein 2 (M2) is a highly conserved 97 amino acid long transmembrane tetrameric protein present in the envelope of IAV. More than 99 % of IAV strains circulating in American swine herds share the identical pandemic (pdm) isoform of M2, making it an ideal target antigen for a vaccine that could elicit broadly protective immunity. Here, using soluble nanoscale membrane assemblies termed nanodiscs (NDs), we designed this membrane mimetic nanostructures displaying full-length M2 in its natural transmembrane configuration (M2ND). Intramuscular (IM) immunization of swine with M2ND mixed with conventional emulsion adjuvant elicited M2-specific IgG antibodies in the serum that recognized influenza virions and M2-specific interferon-γ secreting cells present in the blood. Intranasal (IN) immunization with M2ND adjuvanted with a mycobacterial extract elicited M2-specific IgA in mucosal secretions that also recognized IAV. Immunization with an influenza whole inactivated virus (WIV) vaccine supplemented with a concurrent IM injection of M2ND mixed with an emulsion adjuvant increased the level of protective immunity afforded by the former against a challenge with an antigenically distinct H3N2 IAV, as exhibited by an enhanced elimination of virus from the lung. The lone IM administration of the M2ND vaccine mixed with an emulsion adjuvant provided measurable protection as evidenced by a >10-fold reduction or complete elimination of the challenge virus from the lung, but it did not diminish the viral load in nasal secretions nor the extent of pneumonia that ensued after the virus challenge. In contrast, an improved formulation of the M2ND vaccine that incorporated synthetic CpG oligodeoxynucleotides (CpG-ODN) in the nanostructures administered alone, via the IN and IM routes combined, provided a significant level of protective immunity against IAV as evidenced by a decreased viral load in both the upper and lower respiratory tracts and fully eliminated the occurrence of pneumonia in 89 % of the pigs immunized with this biologic. Notably, to be effective, the M2 protein must be displayed in the ND assemblies, as shown by the observation that simply mixing M2 with empty NDs incorporating CpG-ODN (eND-CpG-ODN) did not provide protective immunity. This novel M2-based vaccine offers great promise to help increase the breadth of protection afforded by conventional WIV vaccines against the diversity of IAV in circulation and, plausibly, as a broadly protective stand-alone biologic.
Collapse
Affiliation(s)
- Federico A Zuckermann
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA.
| | - Yelena V Grinkova
- Department of Biochemistry, 505 South Goodwin Avenue, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Robert J Husmann
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Melissa Pires-Alves
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Suzanna Storms
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Wei-Yu Chen
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Stephen G Sligar
- Department of Biochemistry, 505 South Goodwin Avenue, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
25
|
Rider PJF, Dulin H, Uche IK, McGee MC, Huang W, Kousoulas KG, Hai R. A Herpes Simplex Virus Type-1-Derived Influenza Vaccine Induces Balanced Adaptive Immune Responses and Protects Mice From Lethal Influenza Virus Challenge. J Med Virol 2024; 96:e70067. [PMID: 39568407 DOI: 10.1002/jmv.70067] [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: 08/30/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
Influenza virus is a major respiratory viral pathogen responsible for the deaths of hundreds of thousands worldwide each year. Current vaccines provide protection primarily by inducing strain-specific antibody responses with the requirement of a match between vaccine strains and circulating strains. It has been suggested that anti-influenza T-cell responses, in addition to antibody responses may provide the broadest protection against different flu strains. Therefore, to address this urgent need, it is desirable to develop a vaccine candidate with an ability to induce balanced adaptive immunity including cell mediated immune responses. Here, we explored the potential of VC2, a well-characterized Herpes Simplex Virus type 1 vaccine vector, as a live attenuated influenza vaccine candidate. We generated a recombinant VC2 virus expressing the influenza A hemagglutinin protein. We show that this virus is capable of generating potent and specific anti-influenza humoral and cell-mediated immune responses. We further show that a single vaccination with the VC2-derived influenza vaccine protects mice from lethal challenge with influenza virus. Our data support the continued development of VC2-derived influenza vaccines for protection of human populations from both seasonal and pandemic strains of influenza. Finally, our results support the potential of VC2-derived vaccines as a platform for the rapid development of vaccines against emerging and established pathogens, particularly respiratory pathogens.
Collapse
MESH Headings
- Animals
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Adaptive Immunity
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Mice
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Mice, Inbred BALB C
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Immunity, Cellular
- Disease Models, Animal
- Humans
- Survival Analysis
- Genetic Vectors/immunology
Collapse
Affiliation(s)
- Paul J F Rider
- Division of Biotechnology and Molecular Medicine Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Harrison Dulin
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| | - Ifeanyi K Uche
- Division of Biotechnology and Molecular Medicine Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Michael C McGee
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Konstantin G Kousoulas
- Division of Biotechnology and Molecular Medicine Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Rong Hai
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, USA
| |
Collapse
|
26
|
Cobey S. Vaccination against rapidly evolving pathogens and the entanglements of memory. Nat Immunol 2024; 25:2015-2023. [PMID: 39384979 DOI: 10.1038/s41590-024-01970-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/26/2024] [Indexed: 10/11/2024]
Abstract
Immune memory determines infection risk and responses to future infections and vaccinations over potentially decades of life. Despite its centrality, the dynamics of memory to antigenically variable pathogens remains poorly understood. This Review examines how past exposures shape B cell responses to vaccinations with influenza and SARS-CoV-2. An overriding feature of vaccinations with these pathogens is the recall of primary responses, often termed 'imprinting' or 'original antigenic sin'. These recalled responses can inhibit the generation of new responses unless some incompletely defined conditions are met. Depending on the context, immune memory can increase or decrease the total neutralizing antibody response to variant antigens, with apparent consequences for protection. These effects are easier to measure experimentally than epidemiologically, but there is evidence that both early and recent exposures influence vaccine effectiveness. A few immunological interactions between adaptive immune responses and antigens might explain the seemingly discrepant effects of memory. Overall, the complex observations point to a need for more quantitative approaches to integrate high-dimensional immune data from populations with diverse exposure histories. Such approaches could help identify optimal vaccination strategies against antigenically diverse pathogens.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
27
|
Feoktistova SG, Ivanova AO, Degtyarev EP, Smirnova DI, Volchkov PY, Deviatkin AA. Phylogenetic Insights into H7Nx Influenza Viruses: Uncovering Reassortment Patterns and Geographic Variability. Viruses 2024; 16:1656. [PMID: 39599771 PMCID: PMC11598867 DOI: 10.3390/v16111656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Influenza A viruses (IAVs), which belong to the Orthomyxoviridae family, are RNA viruses characterized by a segmented genome that allows them to evolve and adapt rapidly. These viruses are mainly transmitted by wild waterfowl. In this study, we investigated the evolutionary processes of H7Nx (H7N1, H7N2, H7N3, H7N4, H7N5, H7N6, H7N7, H7N8, H7N9) viruses, which pose a significant pandemic risk due to the known cases of human infection and their potential for rapid genetic evolution and reassortment. The complete genome sequences of H7Nx influenza viruses (n = 3239) were compared between each other to investigate their phylogenetic relationships and reassortment patterns. For the selected viruses, phylogenetic trees were constructed for eight genome segments (PB2, PB1, PA, HA, NP, NA, M, NS) to assess the genetic diversity and geographic distribution of these viruses. Distinct phylogenetic clades with remarkable geographic patterns were found for the different segments. While the viruses were consistently grouped by subtype based on the NA segment sequences, the phylogeny of the other segment sequences, with the exception of the NS segment, showed distinct grouping patterns based on geographic origin rather than formal subtype assignment. Reassortment events leading to complex phylogenetic relationships were frequently observed. In addition, multiple cases of previously undescribed reassortments between subtypes were detected, emphasizing the fluidity of H7Nx virus populations. These results indicate a high degree of genetic diversity and reassortment within H7Nx influenza viruses. In other words, H7Nx viruses exist as constantly changing combinations of gene pools rather than stable genetic lineages.
Collapse
Affiliation(s)
- Sofya G. Feoktistova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.Y.V.)
| | - Alexandra O. Ivanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, RAS (IBCh RAS), 117997 Moscow, Russia
| | - Egor P. Degtyarev
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.Y.V.)
| | - Daria I. Smirnova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.Y.V.)
| | - Pavel Yu. Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.Y.V.)
- Center for Personalized Medicine, The MCSC Named After A.S. Loginov, 111123 Moscow, Russia
| | - Andrei A. Deviatkin
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.Y.V.)
| |
Collapse
|
28
|
Matz HC, Yu TG, Zhou JQ, Peyton L, Madsen A, Han F, Schmitz AJ, Horvath SC, Dixit K, Keplinger HK, Strnad BS, Hoegger MJ, Middleton WD, Klebert MK, Lin NH, Nachbagauer R, Paris R, Turner JS, Presti RM, Lee J, Ellebedy AH. mRNA-based influenza vaccine expands breadth of B cell response in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617255. [PMID: 39416092 PMCID: PMC11483064 DOI: 10.1101/2024.10.10.617255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Eliciting broad and durable antibody responses against rapidly evolving pathogens like influenza viruses remains a formidable challenge1,2. The germinal center (GC) reaction enables the immune system to generate broad, high-affinity, and durable antibody responses to vaccination3-5. mRNA-based severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines induce persistent GC B cell responses in humans6-9. Whether an mRNA-based influenza vaccine could induce a superior GC response in humans compared to the conventional inactivated influenza virus vaccine remains unclear. We assessed B cell responses in peripheral blood and draining lymph nodes in cohorts receiving the inactivated or mRNA-based quadrivalent seasonal influenza vaccine. Participants receiving the mRNA-based vaccine produced more robust plasmablast responses and higher antibody titers to H1N1 and H3N2 influenza A viruses and comparable antibody titers against influenza B virus strains. Importantly, mRNA-based vaccination stimulated robust recall B cell responses characterized by sustained GC reactions that lasted at least 26 weeks post-vaccination in three of six participants analyzed. In addition to promoting the maturation of responding B cell clones, these sustained GC reactions resulted in enhanced engagement of low-frequency pre-existing memory B cells, expanding the landscape of vaccine-elicited B cell clones. This translated to expansion of the serological repertoire and increased breadth of serum antibody responses. These findings reveal an important role for the induction of persistent GC responses to influenza vaccination in humans to broaden the repertoire of vaccine-induced antibodies.
Collapse
Affiliation(s)
- Hanover C. Matz
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Tae-Geun Yu
- Thayer School of Engineering, Dartmouth College; Hanover, NH 03755, USA
| | - Julian Q. Zhou
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Lowrey Peyton
- Quantitative Biomedical Sciences Program, Dartmouth College; Lebanon, NH 03756, USA
| | - Anders Madsen
- Influenza Centre, Department of Clinical Science, University of Bergen; 5021 Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, 5009 Bergen, Norway
| | - Fangjie Han
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Aaron J. Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Stephen C. Horvath
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Kritika Dixit
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Hunter K. Keplinger
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Benjamin S. Strnad
- Mallinckrodt Institute of Radiology, Washington University School of Medicine; St Louis, MO 63110, USA
| | - Mark J. Hoegger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine; St Louis, MO 63110, USA
| | - William D. Middleton
- Mallinckrodt Institute of Radiology, Washington University School of Medicine; St Louis, MO 63110, USA
| | - Michael K. Klebert
- Infectious Disease Clinical Research Unit, Washington University School of Medicine; St Louis, MO 63110, USA
| | | | | | | | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Rachel M. Presti
- Infectious Disease Clinical Research Unit, Washington University School of Medicine; St Louis, MO 63110, USA
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine; St Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine; St. Louis, MO 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine; St. Louis, MO 63110, USA
| | - Jiwon Lee
- Thayer School of Engineering, Dartmouth College; Hanover, NH 03755, USA
- Quantitative Biomedical Sciences Program, Dartmouth College; Lebanon, NH 03756, USA
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine; St. Louis, MO 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine; St. Louis, MO 63110, USA
| |
Collapse
|
29
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
30
|
Uno N, Ebensen T, Guzman CA, Ross TM. Intranasal administration of octavalent next-generation influenza vaccine elicits protective immune responses against seasonal and pre-pandemic viruses. J Virol 2024; 98:e0035424. [PMID: 39171925 PMCID: PMC11406897 DOI: 10.1128/jvi.00354-24] [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: 02/23/2024] [Accepted: 06/27/2024] [Indexed: 08/23/2024] Open
Abstract
Development of next-generation influenza virus vaccines is crucial to improve protection against circulating and emerging viruses. Current vaccine formulations have to be updated annually due to mutations in seasonal strains and do not offer protection against strains with pandemic potential. Computationally optimized broadly reactive antigen (COBRA) methodology has been utilized by our group to generate broadly reactive immunogens for individual influenza subtypes, which elicit protective immune responses against a broad range of strains over numerous seasons. Octavalent mixtures of COBRA hemagglutinin (HA) (H1, H2, H3, H5, H7, and influenza B virus) plus neuraminidase (NA) (N1 and N2) recombinant proteins mixed with c-di-AMP adjuvant were administered intranasally to naive or pre-immune ferrets in prime-boost fashion. Four weeks after final vaccination, collected sera were analyzed for breadth of antibody response, and the animals were challenged with seasonal or pre-pandemic strains. The octavalent COBRA vaccine elicited antibodies that recognized a broad panel of strains representing different subtypes, and these vaccinated animals were protected against influenza virus challenges. Overall, this study demonstrated that the mixture of eight COBRA HA/NA proteins mixed with an intranasal adjuvant is a promising candidate for a universal influenza vaccine. IMPORTANCE Influenza is a respiratory virus which infects around a billion people globally every year, with millions experiencing severe illness. Commercial vaccine efficacy varies year to year and can be low due to mismatch of circulating virus strains. Thus, the formulation of current vaccines has to be adapted accordingly every year. The development of a broadly reactive influenza vaccine would lessen the global economic and public health burden caused by the different types of influenza viruses. The significance of our research is producing a promising universal vaccine candidate which provides protection against a wider range of virus strains over a wider range of time.
Collapse
Affiliation(s)
- Naoko Uno
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A Guzman
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, Florida, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
31
|
Rathnasinghe R, Chang LA, Pearl R, Jangra S, Aspelund A, Hoag A, Yildiz S, Mena I, Sun W, Loganathan M, Crossland NA, Gertje HP, Tseng AE, Aslam S, Albrecht RA, Palese P, Krammer F, Schotsaert M, Muster T, García-Sastre A. Sequential immunization with chimeric hemagglutinin ΔNS1 attenuated influenza vaccines induces broad humoral and cellular immunity. NPJ Vaccines 2024; 9:169. [PMID: 39300090 DOI: 10.1038/s41541-024-00952-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
Influenza viruses pose a threat to public health as evidenced by severe morbidity and mortality in humans on a yearly basis. Given the constant changes in the viral glycoproteins owing to antigenic drift, seasonal influenza vaccines need to be updated periodically and effectiveness often drops due to mismatches between vaccine and circulating strains. In addition, seasonal influenza vaccines are not protective against antigenically shifted influenza viruses with pandemic potential. Here, we have developed a highly immunogenic vaccination regimen based on live-attenuated influenza vaccines (LAIVs) comprised of an attenuated virus backbone lacking non-structural protein 1 (ΔNS1), the primary host interferon antagonist of influenza viruses, with chimeric hemagglutinins (cHA) composed of exotic avian head domains with a highly conserved stalk domain, to redirect the humoral response towards the HA stalk. In this study, we showed that cHA-LAIV vaccines induce robust serum and mucosal responses against group 1 stalk and confer antibody-dependent cell cytotoxicity activity. Mice that intranasally received cH8/1-ΔNS1 followed by a cH11/1-ΔNS1 heterologous booster had robust humoral responses for influenza A virus group 1 HAs and were protected from seasonal H1N1 influenza virus and heterologous highly pathogenic avian H5N1 lethal challenges. When compared with mice immunized with the standard of care or cold-adapted cHA-LAIV, cHA-ΔNS1 immunized mice had robust antigen-specific CD8+ T-cell responses which also correlated with markedly reduced lung pathology post-challenge. These observations support the development of a trivalent universal influenza vaccine for the protection against group 1 and group 2 influenza A viruses and influenza B viruses.
Collapse
Affiliation(s)
- Raveen Rathnasinghe
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rebecca Pearl
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Amy Aspelund
- Vivaldi Biosciences Inc., Fort Collins, CO, 80523, USA
| | - Alaura Hoag
- Vivaldi Biosciences Inc., Fort Collins, CO, 80523, USA
| | - Soner Yildiz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Madhumathi Loganathan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicholas Alexander Crossland
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Hans P Gertje
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
| | - Anna Elise Tseng
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sadaf Aslam
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Thomas Muster
- Vivaldi Biosciences Inc., Fort Collins, CO, 80523, USA
- Department of Dermatology, University of Vienna Medical School, 1090, Wien, Austria
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
32
|
D’Onofrio V, Porrez S, Jacobs B, Alhatemi A, De Boever F, Waerlop G, Michels E, Vanni F, Manenti A, Leroux-Roels G, Platenburg PP, Hilgers L, Leroux-Roels I. Safety and Immunogenicity of a Carbohydrate Fatty Acid Monosulphate Ester Adjuvant Combined with a Low-Dose Quadrivalent Split-Virion Inactivated Influenza Vaccine: A Randomised, Observer-Blind, Active-Controlled, First-in-Human, Phase 1 Study. Vaccines (Basel) 2024; 12:1036. [PMID: 39340066 PMCID: PMC11435821 DOI: 10.3390/vaccines12091036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Seasonal influenza vaccine effectiveness is low. Carbohydrate fatty acid monosulphate ester (CMS), a new oil-in-water adjuvant, has proven potency in animal models with suggested capacity for dose-sparing. The objective was to evaluate safety and immunogenicity of CMS when added to a low-dose influenza vaccine (QIV) in humans. In a randomised, double-blind, active-controlled, first-in-human study, sixty participants (18-50 years) received either 0.5 mg CMS or 2 mg CMS with 1/5th dose QIV, or a full dose QIV without CMS. Adverse events (AE) were monitored until 7 days post-vaccination. Haemagglutinin inhibition (HI) titres in serum and CD4+ T cells in PBMCs were determined at day 0, 7, 28, and 180. Mean age was 37.6 (±10.1) years and 42/60 (70.0%) were female. Pain at injection site (42/60, 86.7%) and headache (34/60, 56.7%) were reported most and more frequently in the 2 mg CMS group. HI titres and the frequency of influenza specific CD4+ T cells were equal across strains for the three cohorts on all visits, increased until day 28 and decreased at day 180 to values higher than baseline. CMS was safe in humans. Humoral and cell-mediated immunogenicity was similar across vaccines, even with 1/5th antigen dose. CMS can have beneficial implications in low-resource settings or in a pandemic context.
Collapse
Affiliation(s)
- Valentino D’Onofrio
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Sharon Porrez
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Bart Jacobs
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Azhar Alhatemi
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Fien De Boever
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Gwenn Waerlop
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | - Els Michels
- Harmony Clinical Research BV, 9090 Melle, Belgium;
| | - Francesca Vanni
- VisMederi S.r.l., 53035 Monteriggioni, Italy; (F.V.); (A.M.)
| | | | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| | | | - Luuk Hilgers
- LiteVax, 4061 BJ Ophemert, The Netherlands; (P.P.P.); (L.H.)
| | - Isabel Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University and Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium; (V.D.); (S.P.); (B.J.); (A.A.); (F.D.B.); (G.W.); (G.L.-R.)
| |
Collapse
|
33
|
Ge P, Ross TM. COBRA HA and NA vaccination elicits long-live protective immune responses against pre-pandemic H2, H5, and H7 influenza virus subtypes. Virology 2024; 597:110119. [PMID: 38850895 DOI: 10.1016/j.virol.2024.110119] [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: 03/19/2024] [Revised: 05/08/2024] [Accepted: 05/21/2024] [Indexed: 06/10/2024]
Abstract
Highly pathogenic avian influenza (HPAI) viruses remain a major threat to both the poultry industry and human public health, and these viruses continue to spread worldwide. In this study, mice were vaccinated with COBRA H2, H5, and H7 hemagglutinin (HA) and two neuraminidase (NA) proteins, N1 and N2. Vaccinated mice were fully protected against lethal challenge with H5N6 influenza virus. Sera collected after vaccination showed cross-reactive IgG antibodies against a panel of wild-type H2, H5, and H7 HA proteins, and N1 and N2 NA proteins. Mice with pre-existing immunity to H1N1 and H3N2 influenza viruses that were subsequently vaccinated with COBRA HA/NA vaccines had enhanced anti-HA stem antibodies compared to vaccinated mice without pre-existing immunity. In addition, sera collected after vaccination had hemagglutinin inhibitory activity against a panel of H2Nx, H5Nx, and H7Nx influenza viruses. These protective antibodies were maintained up for up to 4 months after vaccination.
Collapse
Affiliation(s)
- Pan Ge
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; 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; Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, USA; Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
34
|
Cowling BJ, Okoli GN. Influenza Vaccine Effectiveness and Progress Towards a Universal Influenza Vaccine. Drugs 2024; 84:1013-1023. [PMID: 39167316 PMCID: PMC11438668 DOI: 10.1007/s40265-024-02083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2024] [Indexed: 08/23/2024]
Abstract
At various times in recent decades, surges have occurred in optimism about the potential for universal influenza vaccines that provide strong, broad, and long-lasting protection and could substantially reduce the disease burden associated with seasonal influenza epidemics as well as the threat posed by pandemic influenza. Each year more than 500 million doses of seasonal influenza vaccine are administered around the world, with most doses being egg-grown inactivated subunit or split-virion vaccines. These vaccines tend to have moderate effectiveness against medically attended influenza for influenza A(H1N1) and influenza B, and somewhat lower for influenza A(H3N2) where differences between vaccine strains and circulating strains can occur more frequently due to antigenic drift and egg adaptations in the vaccine strains. Several enhanced influenza vaccine platforms have been developed including cell-grown antigen, the inclusion of adjuvants, or higher antigen doses, to improve immunogenicity and protection. During the COVID-19 pandemic there was unprecedented speed in development and roll-out of relatively new vaccine platforms, including mRNA vaccines and viral vector vaccines. These new platforms present opportunities to improve protection for influenza beyond existing products. Other approaches continue to be explored. Incremental improvements in influenza vaccine performance should be achievable in the short to medium term.
Collapse
Affiliation(s)
- Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, Hong Kong, China.
| | - George N Okoli
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
35
|
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
|
36
|
Xu D, Carter JJ, Li C, Utz A, Weidenbacher PAB, Tang S, Sanyal M, Pulendran B, Barnes CO, Kim PS. Vaccine design via antigen reorientation. Nat Chem Biol 2024; 20:1012-1021. [PMID: 38225471 PMCID: PMC11247139 DOI: 10.1038/s41589-023-01529-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024]
Abstract
A major challenge in creating universal influenza vaccines is to focus immune responses away from the immunodominant, variable head region of hemagglutinin (HA-head) and toward the evolutionarily conserved stem region (HA-stem). Here we introduce an approach to control antigen orientation via site-specific insertion of aspartate residues that facilitates antigen binding to alum. We demonstrate the generalizability of this approach with antigens from Ebola, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses and observe enhanced neutralizing antibody responses in all cases. We then reorient an H2 HA in an 'upside-down' configuration to increase the exposure and immunogenicity of HA-stem. The reoriented H2 HA (reoH2HA) on alum induced stem-directed antibodies that cross-react with both group 1 and group 2 influenza A subtypes. Electron microscopy polyclonal epitope mapping (EMPEM) revealed that reoH2HA (group 1) elicits cross-reactive antibodies targeting group 2 HA-stems. Our results highlight antigen reorientation as a generalizable approach for designing epitope-focused vaccines.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Joshua J Carter
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton A B Weidenbacher
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Shaogeng Tang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher O Barnes
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Peter S Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
37
|
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
|
38
|
Bufan B, Arsenović-Ranin N, Živković I, Ćuruvija I, Blagojević V, Dragačević L, Kovačević A, Kotur-Stevuljević J, Leposavić G. Modulation of T-Cell-Dependent Humoral Immune Response to Influenza Vaccine by Multiple Antioxidant/Immunomodulatory Micronutrient Supplementation. Vaccines (Basel) 2024; 12:743. [PMID: 39066381 PMCID: PMC11281378 DOI: 10.3390/vaccines12070743] [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: 06/10/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Notwithstanding prevalence gaps in micronutrients supporting immune functions, the significance of their deficits/supplementation for the efficacy of vaccines is underinvestigated. Thus, the influence of supplementation combining vitamins C and D, zinc, selenium, manganese, and N-acetyl cysteine on immune correlates/surrogates of protection conferred by a quadrivalent influenza vaccine (QIV) in mice was investigated. The supplementation starting 5 days before the first of two QIV injections given 28 days apart increased the serum titres of total and neutralizing IgG against each of four influenza strains from QIV. Accordingly, the frequencies of germinal center B cells, follicular CD4+ T helper (Th) cells, and IL-21-producing Th cells increased in secondary lymphoid organs (SLOs). Additionally, the supplementation improved already increased IgG response to the second QIV injection by augmenting not only neutralizing antibody production, but also IgG2a response, which is important for virus clearance, through favoring Th1 differentiation as indicated by Th1 (IFN-γ)/Th2 (IL-4) signature cytokine level ratio upon QIV restimulation in SLO cell cultures. This most likely partly reflected antioxidant action of the supplement as indicated by splenic redox status analyses. Thus, the study provides a solid scientific background for further research aimed at repurposing the use of this safe and inexpensive micronutrient combination to improve response to the influenza vaccine.
Collapse
Affiliation(s)
- Biljana Bufan
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (B.B.); (N.A.-R.)
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (B.B.); (N.A.-R.)
| | - Irena Živković
- Department of Research and Development, Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ž.); (I.Ć.); (V.B.); (L.D.)
| | - Ivana Ćuruvija
- Department of Research and Development, Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ž.); (I.Ć.); (V.B.); (L.D.)
| | - Veljko Blagojević
- Department of Research and Development, Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ž.); (I.Ć.); (V.B.); (L.D.)
| | - Luka Dragačević
- Department of Research and Development, Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ž.); (I.Ć.); (V.B.); (L.D.)
| | - Ana Kovačević
- Department for Virology Control, Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia;
| | - Jelena Kotur-Stevuljević
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia;
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia
| |
Collapse
|
39
|
Simmons HC, Finney J, Kotaki R, Adachi Y, Park Moseman A, Watanabe A, Song S, Robinson-McCarthy LR, Le Sage V, Kuraoka M, Moseman EA, Kelsoe G, Takahashi Y, McCarthy KR. A protective and broadly binding antibody class engages the influenza virus hemagglutinin head at its stem interface. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.13.571543. [PMID: 38168412 PMCID: PMC10760138 DOI: 10.1101/2023.12.13.571543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Influenza infection and vaccination impart strain-specific immunity that protects against neither seasonal antigenic variants nor the next pandemic. However, antibodies directed to conserved sites can confer broad protection. Here we identify and characterize a class of human antibodies that engage a previously undescribed, conserved epitope on the influenza hemagglutinin (HA) protein. Prototype antibody S8V1-157 binds at the normally occluded interface between the HA head and stem. Antibodies to this HA head-stem interface epitope are non-neutralizing in vitro but protect against lethal influenza infection in mice. Antibody isotypes that direct clearance of infected cells enhance this protection. Head-stem interface antibodies bind to most influenza A serotypes and seasonal human variants, and are present at low frequencies in the memory B cell populations of multiple human donors. Vaccines designed to elicit these antibodies might contribute to "universal" influenza immunity.
Collapse
Affiliation(s)
- Holly C. Simmons
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joel Finney
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Ryutaro Kotaki
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yu Adachi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Annie Park Moseman
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Akiko Watanabe
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Shengli Song
- Department of Surgery, Duke University, Durham, North Carolina 27710, USA
| | - Lindsey R. Robinson-McCarthy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Valerie Le Sage
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Masayuki Kuraoka
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - E. Ashley Moseman
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Garnett Kelsoe
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kevin R. McCarthy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Hammershaimb EAD, Campbell JD. Vaccine Development. Pediatr Clin North Am 2024; 71:529-549. [PMID: 38754940 DOI: 10.1016/j.pcl.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
This article considers ethical considerations surrounding pediatric vaccine development for pandemic preparedness, examines some historical cases of pediatric vaccines developed during past smallpox, influenza, and 2019 coronavirus disease pandemics, and discusses the current state of vaccine development for pandemic preparedness, including vaccines against smallpox/mpox, influenza, anthrax, and Ebola that are included in the US Strategic National Stockpile and vaccines being developed against priority pathogens identified by the World Health Organization.
Collapse
Affiliation(s)
- Elizabeth A D Hammershaimb
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Room 480, Baltimore, MD 21201, USA; Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - James D Campbell
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Room 480, Baltimore, MD 21201, USA; Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Lopez CE, Zacharias ZR, Ross KA, Narasimhan B, Waldschmidt TJ, Legge KL. Polyanhydride nanovaccine against H3N2 influenza A virus generates mucosal resident and systemic immunity promoting protection. NPJ Vaccines 2024; 9:96. [PMID: 38822003 PMCID: PMC11143372 DOI: 10.1038/s41541-024-00883-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 05/07/2024] [Indexed: 06/02/2024] Open
Abstract
Influenza A virus (IAV) causes significant morbidity and mortality worldwide due to seasonal epidemics and periodic pandemics. The antigenic drift/shift of IAV continually gives rise to new strains and subtypes, aiding IAV in circumventing previously established immunity. As a result, there has been substantial interest in developing a broadly protective IAV vaccine that induces, durable immunity against multiple IAVs. Previously, a polyanhydride nanoparticle-based vaccine or nanovaccine (IAV-nanovax) encapsulating H1N1 IAV antigens was reported, which induced pulmonary B and T cell immunity and resulted in cross-strain protection against IAV. A key feature of IAV-nanovax is its ability to easily incorporate diverse proteins/payloads, potentially increasing its ability to provide broad protection against IAV and/or other pathogens. Due to human susceptibility to both H1N1 and H3N2 IAV, several H3N2 nanovaccines were formulated herein with multiple IAV antigens to examine the "plug-and-play" nature of the polyanhydride nanovaccine platform and determine their ability to induce humoral and cellular immunity and broad-based protection similar to IAV-nanovax. The H3N2-based IAV nanovaccine formulations induced systemic and mucosal B cell responses which were associated with antigen-specific antibodies. Additionally, systemic and lung-tissue resident CD4 and CD8 T cell responses were enhanced post-vaccination. These immune responses corresponded with protection against both homologous and heterosubtypic IAV infection. Overall, these results demonstrate the plug-and-play nature of the polyanhydride nanovaccine platform and its ability to generate immunity and protection against IAV utilizing diverse antigenic payloads.
Collapse
Affiliation(s)
- Christopher E Lopez
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Zeb R Zacharias
- Interdisciplinary Immunology Graduate Program, Department of Pathology, University of Iowa, Iowa City, IA, USA
| | | | - Balaji Narasimhan
- Nanovaccine Institute, Iowa State University, Ames, IA, USA
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Thomas J Waldschmidt
- Interdisciplinary Immunology Graduate Program, Department of Pathology, University of Iowa, Iowa City, IA, USA
- Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Kevin L Legge
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA.
- Interdisciplinary Immunology Graduate Program, Department of Pathology, University of Iowa, Iowa City, IA, USA.
- Nanovaccine Institute, Iowa State University, Ames, IA, USA.
| |
Collapse
|
42
|
Hill BD, Zak AJ, Raja S, Bugada LF, Rizvi SM, Roslan SB, Nguyen HN, Chen J, Jiang H, Ono A, Goldstein DR, Wen F. iGATE analysis improves the interpretability of single-cell immune landscape of influenza infection. JCI Insight 2024; 9:e172140. [PMID: 38814732 PMCID: PMC11383363 DOI: 10.1172/jci.insight.172140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
Influenza poses a persistent health burden worldwide. To design equitable vaccines effective across all demographics, it is essential to better understand how host factors such as genetic background and aging affect the single-cell immune landscape of influenza infection. Cytometry by time-of-flight (CyTOF) represents a promising technique in this pursuit, but interpreting its large, high-dimensional data remains difficult. We have developed a new analytical approach, in silico gating annotating training elucidating (iGATE), based on probabilistic support vector machine classification. By rapidly and accurately "gating" tens of millions of cells in silico into user-defined types, iGATE enabled us to track 25 canonical immune cell types in mouse lung over the course of influenza infection. Applying iGATE to study effects of host genetic background, we show that the lower survival of C57BL/6 mice compared with BALB/c was associated with a more rapid accumulation of inflammatory cell types and decreased IL-10 expression. Furthermore, we demonstrate that the most prominent effect of aging is a defective T cell response, reducing survival of aged mice. Finally, iGATE reveals that the 25 canonical immune cell types exhibited differential influenza infection susceptibility and replication permissiveness in vivo, but neither property varied with host genotype or aging. The software is available at https://github.com/UmichWenLab/iGATE.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Judy Chen
- Program in Immunology
- Department of Internal Medicine
| | | | - Akira Ono
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Fei Wen
- Department of Chemical Engineering
| |
Collapse
|
43
|
Gao Y, Han S, Lu F, Liu Q, Yang J, Wang W, Wang Y, Zhang J, Ju R, Shen X, Zhao Y, Wang H, Tan W, Wang L. Dimethyl-Dioctadecyl-Ammonium Bromide/Poly(lactic acid) Nanoadjuvant Enhances the Immunity and Cross-Protection of an NM2e-Based Universal Influenza Vaccine. ACS NANO 2024; 18:12905-12916. [PMID: 38721835 DOI: 10.1021/acsnano.4c00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
For most frequent respiratory viruses, there is an urgent need for a universal influenza vaccine to provide cross-protection against intra- and heterosubtypes. We previously developed an Escherichia coli fusion protein expressed extracellular domain of matrix 2 (M2e) and nucleoprotein, named NM2e, and then combined it with an aluminum adjuvant, forming a universal vaccine. Although NM2e has demonstrated a protective effect against the influenza virus in mice to some extent, further improvement is still needed for the induction of immune responses ensuring adequate cross-protection against influenza. Herein, we fabricated a cationic solid lipid nanoadjuvant using poly(lactic acid) (PLA) and dimethyl-dioctadecyl-ammonium bromide (DDAB) and loaded NM2e to generate an NM2e@DDAB/PLA nanovaccine (Nv). In vitro experiments suggested that bone marrow-derived dendritic cells incubated with Nv exhibited ∼4-fold higher antigen (Ag) uptake than NM2e at 16 h along with efficient activation by NM2e@DDAB/PLA Nv. In vivo experiments revealed that Ag of the Nv group stayed in lymph nodes (LNs) for more than 14 days after initial immunization and DCs in LNs were evidently activated and matured. Furthermore, the Nv primed T and B cells for robust humoral and cellular immune responses after immunization. It also induced a ratio of IgG2a/IgG1 higher than that of NM2e to a considerable extent. Moreover, NM2e@DDAB/PLA Nv quickly restored body weight and improved survival of homo- and heterosubtype influenza challenged mice, and the cross-protection efficiency was over 90%. Collectively, our study demonstrated that NM2e@DDAB/PLA Nv could offer notable protection against homo- and heterosubtype influenza virus challenges, offering the potential for the development of a universal influenza vaccine.
Collapse
Affiliation(s)
- Yuan Gao
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Heilongjiang Provincial Key Laboratory of Ecological Utilization of Forestry-Based Active Substances, College of Chemistry, Chemistry Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China
| | - Shulan Han
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, P.R. China
| | - Funa Lu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, P.R. China
- Basic Medical College, Inner Mongolia Medical University, Hohhot 010010, P.R. China
| | - Qi Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jun Yang
- Beijing Economic-Technological Development Area (BDA), Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing 100176, China
| | - Wenling Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Yuanyuan Wang
- Beijing Institute of Petrochemical Technology, Beijing 102617, P.R. China
| | - Jing Zhang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Ruijun Ju
- Beijing Institute of Petrochemical Technology, Beijing 102617, P.R. China
| | - Xiaoling Shen
- Basic Medical College, Inner Mongolia Medical University, Hohhot 010010, P.R. China
| | - Yanping Zhao
- Beijing Economic-Technological Development Area (BDA), Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing 100176, China
| | - Hongjun Wang
- Beijing Economic-Technological Development Area (BDA), Beijing Tide Pharmaceutical Co., Ltd, No.8 East Rongjing Street, Beijing 100176, China
| | - Wenjie Tan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Lianyan Wang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| |
Collapse
|
44
|
Ray R, Nait Mohamed FA, Maurer DP, Huang J, Alpay BA, Ronsard L, Xie Z, Han J, Fernandez-Quintero M, Phan QA, Ursin RL, Vu M, Kirsch KH, Prum T, Rosado VC, Bracamonte-Moreno T, Okonkwo V, Bals J, McCarthy C, Nair U, Kanekiyo M, Ward AB, Schmidt AG, Batista FD, Lingwood D. Eliciting a single amino acid change by vaccination generates antibody protection against group 1 and group 2 influenza A viruses. Immunity 2024; 57:1141-1159.e11. [PMID: 38670113 PMCID: PMC11096021 DOI: 10.1016/j.immuni.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024]
Abstract
Broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin (HA) stem of influenza A viruses (IAVs) tend to be effective against either group 1 or group 2 viral diversity. In rarer cases, intergroup protective bnAbs can be generated by human antibody paratopes that accommodate the conserved glycan differences between the group 1 and group 2 stems. We applied germline-engaging nanoparticle immunogens to elicit a class of cross-group bnAbs from physiological precursor frequency within a humanized mouse model. Cross-group protection depended on the presence of the human bnAb precursors within the B cell repertoire, and the vaccine-expanded antibodies enriched for an N55T substitution in the CDRH2 loop, a hallmark of the bnAb class. Structurally, this single mutation introduced a flexible fulcrum to accommodate glycosylation differences and could alone enable cross-group protection. Thus, broad IAV immunity can be expanded from the germline repertoire via minimal antigenic input and an exceptionally simple antibody development pathway.
Collapse
Affiliation(s)
- Rashmi Ray
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Faez Amokrane Nait Mohamed
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA.
| | - Daniel P Maurer
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Jiachen Huang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Berk A Alpay
- Systems, Synthetic, and Quantitative Biology Program, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Larance Ronsard
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Zhenfei Xie
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Julianna Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Monica Fernandez-Quintero
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of General, Inorganic and Theoretical Chemistry, Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82/III, 6020 Innsbruck, Austria
| | - Quynh Anh Phan
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Rebecca L Ursin
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Mya Vu
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Kathrin H Kirsch
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Thavaleak Prum
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Victoria C Rosado
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Thalia Bracamonte-Moreno
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Vintus Okonkwo
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Julia Bals
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Caitlin McCarthy
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Usha Nair
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 40 Convent Drive, Bethesda, MD 20892-3005, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Aaron G Schmidt
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Facundo D Batista
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA; Department of Biology, The Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Daniel Lingwood
- The Ragon Institute of Mass General, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
45
|
Branda F, Ciccozzi M. Navigating Novel Viral Challenges: Understanding, Tracking, and Mitigating Emerging Threats. Microorganisms 2024; 12:807. [PMID: 38674751 PMCID: PMC11051937 DOI: 10.3390/microorganisms12040807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of new viral threats continues to pose significant challenges to global health security [...].
Collapse
|
46
|
Mazunina EP, Gushchin VA, Kleymenov DA, Siniavin AE, Burtseva EI, Shmarov MM, Mukasheva EA, Bykonia EN, Kozlova SR, Evgrafova EA, Zolotar AN, Shidlovskaya EV, Kirillova ES, Krepkaia AS, Usachev EV, Kuznetsova NA, Ivanov IA, Dmitriev SE, Ivanov RA, Logunov DY, Gintsburg AL. Trivalent mRNA vaccine-candidate against seasonal flu with cross-specific humoral immune response. Front Immunol 2024; 15:1381508. [PMID: 38690272 PMCID: PMC11058219 DOI: 10.3389/fimmu.2024.1381508] [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: 02/03/2024] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Abstract
Seasonal influenza remains a serious global health problem, leading to high mortality rates among the elderly and individuals with comorbidities. Vaccination is generally accepted as the most effective strategy for influenza prevention. While current influenza vaccines are effective, they still have limitations, including narrow specificity for certain serological variants, which may result in a mismatch between vaccine antigens and circulating strains. Additionally, the rapid variability of the virus poses challenges in providing extended protection beyond a single season. Therefore, mRNA technology is particularly promising for influenza prevention, as it enables the rapid development of multivalent vaccines and allows for quick updates of their antigenic composition. mRNA vaccines have already proven successful in preventing COVID-19 by eliciting rapid cellular and humoral immune responses. In this study, we present the development of a trivalent mRNA vaccine candidate, evaluate its immunogenicity using the hemagglutination inhibition assay, ELISA, and assess its efficacy in animals. We demonstrate the higher immunogenicity of the mRNA vaccine candidate compared to the inactivated split influenza vaccine and its enhanced ability to generate a cross-specific humoral immune response. These findings highlight the potential mRNA technology in overcoming current limitations of influenza vaccines and hold promise for ensuring greater efficacy in preventing seasonal influenza outbreaks.
Collapse
Affiliation(s)
- Elena P. Mazunina
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir A. Gushchin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Virology, Lomonosov Moscow State University, Moscow, Russia
- Department of Medical Genetics, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Denis A. Kleymenov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrei E. Siniavin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Elena I. Burtseva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Maksim M. Shmarov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Evgenya A. Mukasheva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Evgeniia N. Bykonia
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Sofia R. Kozlova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elina A. Evgrafova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia N. Zolotar
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena V. Shidlovskaya
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena S. Kirillova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiya S. Krepkaia
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Evgeny V. Usachev
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Nadezhda A. Kuznetsova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Igor A. Ivanov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Sergey E. Dmitriev
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Roman A. Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| | - Denis Y. Logunov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander L. Gintsburg
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N. F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
- Infectiology Department, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
47
|
Jia JZ, Cohen CA, Gu H, McLean MR, Varadarajan R, Bhandari N, Peiris M, Leung GM, Poon LLM, Tsang T, Chung AW, Cowling BJ, Leung NHL, Valkenburg SA. Influenza antibody breadth and effector functions are immune correlates from acquisition of pandemic infection of children. Nat Commun 2024; 15:3210. [PMID: 38615070 PMCID: PMC11016072 DOI: 10.1038/s41467-024-47590-0] [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: 06/07/2023] [Accepted: 04/04/2024] [Indexed: 04/15/2024] Open
Abstract
Cross-reactive antibodies with Fc receptor (FcR) effector functions may mitigate pandemic virus impact in the absence of neutralizing antibodies. In this exploratory study, we use serum from a randomized placebo-controlled trial of seasonal trivalent influenza vaccination in children (NCT00792051) conducted at the onset of the 2009 H1N1 pandemic (pH1N1) and monitored for infection. We found that seasonal vaccination increases pH1N1 specific antibodies and FcR effector functions. Furthermore, prospective baseline antibody profiles after seasonal vaccination, prior to pH1N1 infection, show that unvaccinated uninfected children have elevated ADCC effector function, FcγR3a and FcγR2a binding antibodies to multiple pH1N1 proteins, past seasonal and avian (H5, H7 and H9) strains. Whereas, children that became pH1N1 infected after seasonal vaccination have antibodies focussed to seasonal strains without FcR functions, and greater aggregated HA-specific profiles for IgM and IgG3. Modeling to predict infection susceptibility, ranked baseline hemagglutination antibody inhibition as the highest contributor to lack of pH1N1 infection, in combination with features that include pH1-IgG1, H1-stem responses and FcR binding to seasonal vaccine and pH1 proteins. Thus, seasonal vaccination can have benefits against pandemic influenza viruses, and some children already have broadly reactive antibodies with Fc potential without vaccination and may be considered 'elite influenza controllers'.
Collapse
Affiliation(s)
- Janice Z Jia
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Carolyn A Cohen
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Haogao Gu
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Milla R McLean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - Nisha Bhandari
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Malik Peiris
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
- Centre for Immunology and Infection (C2i), Hong Kong Science and Technology Park, Hong Kong, SAR, China
| | - Gabriel M Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
- Laboratory of Data Discovery for Health Limited, Hong Kong Science and Technology Park, Hong Kong, SAR, China
| | - Leo L M Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
- Centre for Immunology and Infection (C2i), Hong Kong Science and Technology Park, Hong Kong, SAR, China
| | - Tim Tsang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Nancy H L Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
48
|
Nie J, Wang Q, Li C, Zhou Y, Yao X, Xu L, Chang Y, Ding F, Sun L, Zhan L, Zhu L, Xie K, Wang X, Shi Y, Zhao Q, Shan Y. Self-Assembled Multiepitope Nanovaccine Provides Long-Lasting Cross-Protection against Influenza Virus. Adv Healthc Mater 2024; 13:e2303531. [PMID: 37983728 DOI: 10.1002/adhm.202303531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Indexed: 11/22/2023]
Abstract
Seasonal influenza vaccines typically provide strain-specific protection and are reformulated annually, which is a complex and time-consuming process. Multiepitope vaccines, combining multiple conserved antigenic epitopes from a pathogen, can trigger more robust, diverse, and effective immune responses, providing a potential solution. However, their practical application is hindered by low immunogenicity and short-term effectiveness. In this study, multiple linear epitopes from the conserved stem domain of hemagglutinin and the ectodomain of matrix protein 2 are combined with the Helicobacter pylori ferritin, a stable self-assembled nanoplatform, to develop an influenza multiepitope nanovaccine, named MHF. MHF is prokaryotically expressed in a soluble form and self-assembles into uniform nanoparticles. The subcutaneous immunization of mice with adjuvanted MHF induces cross-reactive neutralizing antibodies, antibody-dependent cell-mediated cytotoxicity, and cellular immunity, offering complete protection against H3N2 as well as partial protection against H1N1. Importantly, the vaccine cargo delivered by ferritin triggers epitope-specific memory B-cell responses, with antibody level persisting for over 6 months post-immunization. These findings indicate that self-assembled multiepitope nanovaccines elicit potent and long-lasting immune responses while significantly reducing the risk of vaccine escape mutants, and offer greater practicality in terms of scalable manufacturing and genetic manipulability, presenting a promising and effective strategy for future vaccine development.
Collapse
Affiliation(s)
- Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, 519000, China
| | - Qingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Chenxi Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Yongfei Zhou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Lipeng Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Yaotian Chang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Fan Ding
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Lulu Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Li Zhan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Lvzhou Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Kunpeng Xie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Xu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Qi Zhao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, 519000, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 519000, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| |
Collapse
|
49
|
Lian YB, Hu MJ, Guo TK, Yang YL, Zhang RR, Huang JS, Yu LJ, Shi CW, Yang GL, Huang HB, Jiang YL, Wang JZ, Cao X, Wang N, Zeng Y, Yang WT, Wang CF. The protective effect of intranasal immunization with influenza virus recombinant adenovirus vaccine on mucosal and systemic immune response. Int Immunopharmacol 2024; 130:111710. [PMID: 38394888 DOI: 10.1016/j.intimp.2024.111710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Influenza virus is a kind of virus that poses several hazards of animal and human health. Therefore, it is important to develop an effective vaccine to prevent influenza. To this end we successfully packaged recombinant adenovirus rAd-NP-M2e-GFP expressing multiple copies of influenza virus conserved antigens NP and M2e and packaged empty vector adenovirus rAd-GFP. The effect of rAd-NP-M2e-GFP on the activation of dendritic cell (DC) in vitro and in vivo was detected by intranasal immunization. The results showed that rAd-NP-M2e-GFP promoted the activation of DC in vitro and in vivo. After the primary immunization and booster immunization of mice through the nasal immune way, the results showed that rAd-NP-M2e-GFP induced enhanced local mucosal-specific T cell responses, increased the content of SIgA in broncho alveolar lavage fluids (BALF) and triggered the differentiation of B cells in the germinal center. It is proved that rAd-NP-M2e-GFP can significantly elicit mucosal immunity and systemic immune response. In addition, rAd-NP-M2e-GFP could effectively protect mice after H1N1 influenza virus challenge. To lay the foundation and provide reference for further development of influenza virus mucosal vaccine in the future.
Collapse
Affiliation(s)
- Yi-Bing Lian
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Man-Jie Hu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Tian-Kui Guo
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yong-Lei Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Rong-Rong Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jing-Shu Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Ling-Jiao Yu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chun-Wei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Gui-Lian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Hai-Bin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yan-Long Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jian-Zhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wen-Tao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Chun-Feng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
50
|
Laszlofy C, Fazekas G, Barath Z, Vajo Z. Evaluation of Vaccine Immunogenicity-Correlates to Real-World Protection: Influenza. Viruses 2024; 16:441. [PMID: 38543806 PMCID: PMC10975834 DOI: 10.3390/v16030441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 05/23/2024] Open
Abstract
Recent events highlighted that, despite decades of studying vaccine immunogenicity and efforts toward finding correlates of protection, evaluating real-world vaccine efficacy as well as establishing meaningful licensing criteria still represents a significant challenge. In this paper, we review all aspects of influenza vaccine immunogenicity, including animal and human challenge studies, humoral and cellular immunity parameters, and their potential correlation with real-life protection from disease.
Collapse
Affiliation(s)
- Csaba Laszlofy
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, 6720 Szeged, Hungary; (C.L.); (Z.B.)
| | | | - Zoltan Barath
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, 6720 Szeged, Hungary; (C.L.); (Z.B.)
| | - Zoltan Vajo
- Department of Family Medicine, Semmelweis University Medical School, 1085 Budapest, Hungary
| |
Collapse
|