1
|
Kandinov B, Soens M, Huang W, Llapur C, Ensz D, Essink B, Fierro C, Vakil J, Pucci A, Guo J, Rudden S, Hall K, Hicks B, Schaefers K, Zhou H, Ma C, Zheng L, Avanesov A, Park Y, Du E, Miller J, Ananworanich J, Nachbagauer R. An mRNA-based seasonal influenza vaccine in adults: Results of two phase 3 randomized clinical trials and correlate of protection analysis of hemagglutination inhibition titers. Hum Vaccin Immunother 2025; 21:2484088. [PMID: 40174609 PMCID: PMC11970786 DOI: 10.1080/21645515.2025.2484088] [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: 01/10/2025] [Revised: 03/05/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025] Open
Abstract
The safety, immunogenicity, and efficacy of the original formulation of the investigational mRNA-1010 vaccine for seasonal influenza were investigated in two randomized, active-controlled, phase 3 trials in adults (NCT05415462 and NCT05566639), and the results were used to evaluate hemagglutination inhibition (HAI) titers as correlates of risk and protection against influenza-like illness. mRNA-1010 (50-µg) demonstrated an acceptable reactogenicity and safety profile among the >14,000 adult participants vaccinated in both trials. The efficacy profile of mRNA-1010 was generally reflective of immunogenicity findings, with higher immune responses against influenza A strains and lower responses against influenza B strains relative to an active comparator (licensed inactivated influenza vaccine). An analysis of HAI titers as a correlate of protection against influenza infection provided support for its use as a surrogate endpoint for mRNA-1010, similar to licensed influenza vaccines. These findings support further optimization and development of mRNA-1010 against seasonal influenza.
Collapse
Affiliation(s)
| | - Mieke Soens
- Development, Moderna Inc, Cambridge, MA, USA
| | | | | | - David Ensz
- Meridian Clinical Research, Sioux City, IA, USA
| | | | | | | | | | - Jia Guo
- Development, Moderna Inc, Cambridge, MA, USA
| | | | - Kristi Hall
- Development, Moderna Inc, Cambridge, MA, USA
| | - Bryony Hicks
- Development, Moderna Biopharma Canada Corporation, Toronto, ON, Canada
| | | | | | - Chong Ma
- Development, Moderna Inc, Cambridge, MA, USA
| | | | | | | | - Evelyn Du
- Development, Moderna Inc, Cambridge, MA, USA
| | | | | | | |
Collapse
|
2
|
Jiao L, Song Z, Zhou Y, Zhu T, Yu R, Wang Z, Qiu Y, Miao J, Zhang S, Liu Z, Wang D. Naringenin as a phytogenic adjuvant systematically enhances the protective efficacy of H9N2 inactivated vaccine through coordinated innate-adaptive immune priming in chickens. Poult Sci 2025; 104:105257. [PMID: 40344923 DOI: 10.1016/j.psj.2025.105257] [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: 03/17/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025] Open
Abstract
Although inactivated vaccines remain the primary strategy for preventing and controlling avian influenza virus, they fail to induce durable and systemic immune protection. Adjuvants are crucial for enhancing antigen immunogenicity and improving immune responses. In this study, we evaluated the adjuvant activity of naringenin (Nar) for H9N2 inactivated vaccine by detecting humoral immunity, cellular immunity, and viral challenge. The results demonstrated that Nar/H9N2 co-administration significantly increased IgG levels and hemagglutination inhibition (HI) titers. Nar/H9N2 promoted the formation of high-affinity antibodies by upregulating the expression of genes associated with B cell activation and germinal centers (GCs) formation, thus facilitating humoral immune responses. Concurrently, Nar/H9N2 vaccine enhanced T cell proliferation, CD4+and CD8+T cell differentiation, and the expression of Th1/Th2 cytokines, thereby promoting cellular immunity. Crucially, compared to the inactivated H9N2 vaccine alone, viral challenge experiments confirmed that Nar-adjuvanted immunization confers superior protection, markedly reducing viral shedding and minimizing damage to the trachea and lungs. These findings elucidate the capacity of naringenin to synchronize multifaceted immune activation through GCs optimization and T-cell modulation, establishing Nar as a viable candidate for poultry vaccine adjuvants.
Collapse
Affiliation(s)
- Lina Jiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zuchen Song
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yantong Zhou
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianyu Zhu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ruihong Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zheng Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yawei Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Shun Zhang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
3
|
Alam MM, Salauddin A, Moni S, Limon MBH, Musarrat R, Bosu S, Hossain ME, Rahman MZ, Rahman M. Cross-neutralization of Influenza A by SARS-CoV-2 specific neutralizing antibodies and polyclonal plasma: Is pre-exposure to SARS-CoV-2 protective against Influenza A? Heliyon 2024; 10:e40638. [PMID: 39654774 PMCID: PMC11626022 DOI: 10.1016/j.heliyon.2024.e40638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
According to sparse information from various countries, the seasonal influenza virus circulation has drastically decreased during the COVID-19 pandemic. Here, we show the cross-reactivity of anti-SARS-CoV-2 antibodies against influenza viruses. Plasma samples were collected from 311 SARS-CoV-2 infected individuals. The samples were tested for antibody titers against SARS-CoV-2 by ELISA and seasonal influenza virus strains (influenza A/H1N1, A/H3N2, B/Yamagata, and B/Victoria) using a Hemagglutination Inhibition Assay (HAI). In addition, SARS-CoV-2 antibody-positive but Influenza antibody-negative samples (n = 16) were investigated to determine the SARS-CoV-2 antibody-neutralizing potential against influenza viruses by microneutralization (MN) assay. The SARS-CoV-2 genomes were sequenced using Illumina next-generation sequencing, and an in-silico protein structural analysis was performed to identify epitope and antibody binding similarities between SARS-CoV-2 and influenza viruses. Among 16 samples that didn't contain antibodies against Influenza A strains (H1N1 and H3N2), five showed high (MN titer≥20), and six showed moderate (MN titer≥10) capability to neutralize Influenza A. Subsequent in-silico analysis revealed that most efficient binding (>8 kcal/mol) was found between the antibodies of SARS-CoV-2 delta variant (ΔG) with influenza A/H1N1 HA (Hemagglutinin), A/H3N2 HA, A/H1N1 NA (Neuraminidase), and A/H3N2 NA glycoproteins with -12.4, -9.3, -10.1, and -11.7 kcal/mol, respectively. This investigation revealed that neutralizing antibodies of the delta variant cross-reacted with the Influenza A virus, which might protect against influenza viruses and reduce and shift the seasonal influenza circulation during the COVID-19 pandemic. Our findings warrant further study to explain the probable mechanisms of this cross-reactivity.
Collapse
Affiliation(s)
- Mohammad Mamun Alam
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Asma Salauddin
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Sayra Moni
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Md Belayet Hasan Limon
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Raisha Musarrat
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Sagar Bosu
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Mohammad Enayet Hossain
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Mohammed Ziaur Rahman
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| | - Mustafizur Rahman
- Virology Laboratory, Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, (icddr,b), Bangladesh
| |
Collapse
|
4
|
Loes AN, Tarabi RAL, Huddleston J, Touyon L, Wong SS, Cheng SMS, Leung NHL, Hannon WW, Bedford T, Cobey S, Cowling BJ, Bloom JD. High-throughput sequencing-based neutralization assay reveals how repeated vaccinations impact titers to recent human H1N1 influenza strains. J Virol 2024; 98:e0068924. [PMID: 39315814 PMCID: PMC11494878 DOI: 10.1128/jvi.00689-24] [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: 04/18/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
The high genetic diversity of influenza viruses means that traditional serological assays have too low throughput to measure serum antibody neutralization titers against all relevant strains. To overcome this challenge, we developed a sequencing-based neutralization assay that simultaneously measures titers against many viral strains using small serum volumes using a workflow similar to traditional neutralization assays. The key innovation is to incorporate unique nucleotide barcodes into the hemagglutinin (HA) genomic segment, and then pool viruses with numerous different barcoded HA variants and quantify the infectivity of all of them simultaneously using next-generation sequencing. With this approach, a single researcher performed the equivalent of 2,880 traditional neutralization assays (80 serum samples against 36 viral strains) in approximately 1 month. We applied the sequencing-based assay to quantify the impact of influenza vaccination on neutralization titers against recent human H1N1 strains for individuals who had or had not also received a vaccine in the previous year. We found that the viral strain specificities of the neutralizing antibodies elicited by vaccination vary among individuals and that vaccination induced a smaller increase in titers for individuals who had also received a vaccine the previous year-although the titers 6 months after vaccination were similar in individuals with and without the previous-year vaccination. We also identified a subset of individuals with low titers to a subclade of recent H1N1 even after vaccination. We provide an experimental protocol (dx.doi.org/10.17504/protocols.io.kqdg3xdmpg25/v1) and computational pipeline (https://github.com/jbloomlab/seqneut-pipeline) for the sequencing-based neutralization assays to facilitate the use of this method by others. IMPORTANCE We describe a new approach that can rapidly measure how the antibodies in human serum inhibit infection by many different influenza strains. This new approach is useful for understanding how viral evolution affects antibody immunity. We apply the approach to study the effect of repeated influenza vaccination.
Collapse
MESH Headings
- Humans
- High-Throughput Nucleotide Sequencing/methods
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Influenza, Human/virology
- Neutralization Tests/methods
- Vaccination
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Adult
- Female
Collapse
Affiliation(s)
- Andrea N Loes
- Howard Hughes Medical Institute, Seattle, Washington, USA
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rosario Araceli L Tarabi
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - John Huddleston
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Lisa Touyon
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Sook San Wong
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Samuel M S Cheng
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Nancy H L Leung
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - William W Hannon
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Trevor Bedford
- Howard Hughes Medical Institute, Seattle, Washington, USA
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois, USA
| | - Benjamin J Cowling
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Jesse D Bloom
- Howard Hughes Medical Institute, Seattle, Washington, USA
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
5
|
Hoen L, Lartey S, Zhou F, Pathirana RD, Krammer F, Mohn KGI, Cox RJ, Brokstad KA. Impact of Pre-Existing Immunity and Age on Antibody Responses to Live Attenuated Influenza Vaccine. Vaccines (Basel) 2024; 12:864. [PMID: 39203990 PMCID: PMC11359048 DOI: 10.3390/vaccines12080864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Live attenuated influenza vaccines (LAIV) typically induce a poor hemagglutination inhibition (HI) response, which is the standard correlate of protection for inactivated influenza vaccines. The significance of the HI response is complicated because the LAIV vaccine primarily induces the local mucosal immune system, while the HI assay measures the circulating serum antibody response. However, age and pre-existing immunity have been identified as important factors affecting LAIV immunogenicity. This study aimed to extend our understanding of LAIV-induced immunity, particularly, the impact age and pre-existing immunity have on eliciting functional and neutralising antibody responses in paediatric and adult populations vaccinated with LAIV. Thirty-one children and 26 adults were immunized with the trivalent LAIV during the 2013-2014 influenza season in Norway. Children under 9 years received a second dose of LAIV 28 days after the first dose. Blood samples were collected pre- and post-vaccination. HI, microneutralization (MN) and enzyme-linked lectin assay for neuraminidase (NA) antibodies were measured against the vaccine strains. IgG antibody avidity against hemagglutinin (HA) and NA proteins from the vaccine strains was also assessed. Significant correlations were observed between HI and MN responses to A/California/7/2009 (A/H1N1)pdm09-like strain and B/Massachusetts/2/2012-like strain, suggesting that MN is a potential immunological correlate for LAIV. However, the relationship between recipient age (or priming status) and serological response varied between vaccine strains. There was a notable increase in HI and MN responses in all cohorts except naive children against the H1N1 strain, where most recipients had responses below the protective antibody threshold. NAI responses were generally weak in naive children against all vaccine strains compared with adults or antigen-primed children. Post-vaccination antibody avidity increased only in primed children below nine years of age against the A/H1N1 strain. Overall, our findings indicate that LAIV elicits functional and neutralizing antibody responses in both naive and antigen experienced cohorts, however, the magnitude and kinetics of the response varies between vaccine strains.
Collapse
Affiliation(s)
- Lukas Hoen
- Influenza Centre, Department of Clinical Science, University of Bergen, Haukelandsbakken, 5009 Bergen, Norway; (S.L.); (F.Z.); (R.D.P.); (K.G.-I.M.); (R.J.C.); (K.A.B.)
| | - Sarah Lartey
- Influenza Centre, Department of Clinical Science, University of Bergen, Haukelandsbakken, 5009 Bergen, Norway; (S.L.); (F.Z.); (R.D.P.); (K.G.-I.M.); (R.J.C.); (K.A.B.)
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Haukelandsbakken, 5009 Bergen, Norway; (S.L.); (F.Z.); (R.D.P.); (K.G.-I.M.); (R.J.C.); (K.A.B.)
| | - Rishi D. Pathirana
- Influenza Centre, Department of Clinical Science, University of Bergen, Haukelandsbakken, 5009 Bergen, Norway; (S.L.); (F.Z.); (R.D.P.); (K.G.-I.M.); (R.J.C.); (K.A.B.)
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- 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, 1090 Vienna, Austria
| | - Kristin G -I Mohn
- Influenza Centre, Department of Clinical Science, University of Bergen, Haukelandsbakken, 5009 Bergen, Norway; (S.L.); (F.Z.); (R.D.P.); (K.G.-I.M.); (R.J.C.); (K.A.B.)
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Rebecca J. Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Haukelandsbakken, 5009 Bergen, Norway; (S.L.); (F.Z.); (R.D.P.); (K.G.-I.M.); (R.J.C.); (K.A.B.)
- Department of Microbiology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Karl A. Brokstad
- Influenza Centre, Department of Clinical Science, University of Bergen, Haukelandsbakken, 5009 Bergen, Norway; (S.L.); (F.Z.); (R.D.P.); (K.G.-I.M.); (R.J.C.); (K.A.B.)
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences (HVL), 5063 Bergen, Norway
| |
Collapse
|
6
|
Garg N, Tellier G, Vale N, Kluge J, Portman JL, Markowska A, Tussey L. Phase 1, randomized, rater and participant blinded placebo-controlled study of the safety, reactogenicity, tolerability and immunogenicity of H1N1 influenza vaccine delivered by VX-103 (a MIMIX microneedle patch [MAP] system) in healthy adults. PLoS One 2024; 19:e0303450. [PMID: 38843267 PMCID: PMC11156369 DOI: 10.1371/journal.pone.0303450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/19/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The MIMIX platform is a novel microneedle array patch (MAP) characterized by slowly dissolving microneedle tips that deploy into the dermis following patch application. We describe safety, reactogenicity, tolerability and immunogenicity for MIMIX MAP vaccination against influenza. METHODOLOGY The trial was a Phase 1, exploratory, first-in-human, parallel randomized, rater, participant, study analyst-blinded, placebo-controlled study in Canada. Forty-five healthy participants (18 to 39 years of age, inclusive) were randomized in a 1:1:1 ratio to receive either 15 μg or 7.5 μg of an H1N1 influenza vaccine, or placebo delivered via MIMIX MAP to the volar forearm. A statistician used a computer program to create a randomization scheme with a block size of 3. Post-treatment follow-up was approximately 180 days. Primary safety outcomes included the incidence of study product related serious adverse events and unsolicited events within 180 days, solicited application site and systemic reactogenicity through 7 days after administration and solicited application site erythema and/or pigmentation 14, 28, 56 and 180 days after administration. Immunogenicity outcomes included antibody titers and percentage of seroconversion (SCR) and seroprotection (SPR) rates determined by the hemagglutination inhibition (HAI) assay. Exploratory outcomes included virus microneutralization (MN) titers, durability and breadth of the immune response. The trial was registered with ClinicalTrials.gov, number NCT06125717. FINDINGS Between July 7, 2022 and March 13, 2023 45 participants were randomized to a treatment group. One participant was lost to follow up in the 15 μg group and 1 participant withdrew from the 7.5 μg dose group. Safety analyses included n = 15 per group, immunogenicity analyses included n = 14 for the 15 μg and 7.5 μg treatment groups and n = 15 for the placebo group. No SAEs were reported in any of the treatment groups. All treatment groups reported solicited local events within 7 days after vaccination, with mild (Grade 1) erythema being the most frequent symptom reported. Other local symptoms reported included mostly mild (Grade 1) induration/swelling, itching, pigmentation, skin flaking, and tenderness. Within 7 days after vaccination, 2 participants (4.4%) reported moderate (Grade 2) erythema, 1 participant (2.2%) reported moderate (Grade 2) induration/swelling, and 1 participant (2.2%) reported moderate (Grade 2) itching. There was an overall reduction in erythema and pigmentation reported on Days 15, 29, 57, and 180 among all treatment groups. Systemic symptoms reported within 7 days after vaccination, included mild (Grade 1) fatigue reported among all treatment groups, and mild (Grade 1) headache reported by 1 participant in the 7.5 μg treatment group. No study drug related severe symptoms were reported in the study. Group mean fold rises in HAI titers ranged between 8.7 and 12-fold, SCRs were >76% and SPRs were >92% for both VX-103 dose groups thereby fulfilling serological criteria established by the EMA and FDA for seasonal influenza vaccines. Longitudinal assessments demonstrate persistence of the immune response through at least Day 180. CONCLUSIONS The MIMIX MAP platform is safe, well tolerated and elicits robust antibody responses.
Collapse
Affiliation(s)
- Naveen Garg
- Centricity Research-Montreal, Point-Claire, Québec, Canada
| | - Guy Tellier
- Centricity Research-Mirabel, Mirabel, Québec, Canada
| | - Noah Vale
- Centricity Research-Toronto, Toronto, Ontario, Canada
| | - Jon Kluge
- Research and Development, Vaxess Technologies, Cambridge, Massachusetts, United States of America
| | - Jonathan L. Portman
- Research and Development, Vaxess Technologies, Cambridge, Massachusetts, United States of America
| | - Anna Markowska
- Research and Development, Vaxess Technologies, Cambridge, Massachusetts, United States of America
| | - Lynda Tussey
- Development and MAP Production, Vaxess Technologies, Woburn, Massachusetts, United States of America
| |
Collapse
|
7
|
Loes AN, Tarabi RAL, Huddleston J, Touyon L, Wong SS, Cheng SMS, Leung NHL, Hannon WW, Bedford T, Cobey S, Cowling BJ, Bloom JD. High-throughput sequencing-based neutralization assay reveals how repeated vaccinations impact titers to recent human H1N1 influenza strains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584176. [PMID: 38496577 PMCID: PMC10942427 DOI: 10.1101/2024.03.08.584176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The high genetic diversity of influenza viruses means that traditional serological assays have too low throughput to measure serum antibody neutralization titers against all relevant strains. To overcome this challenge, we have developed a sequencing-based neutralization assay that simultaneously measures titers against many viral strains using small serum volumes via a workflow similar to traditional neutralization assays. The key innovation is to incorporate unique nucleotide barcodes into the hemagglutinin (HA) genomic segment, and then pool viruses with numerous different barcoded HA variants and quantify infectivity of all of them simultaneously using next-generation sequencing. With this approach, a single researcher performed the equivalent of 2,880 traditional neutralization assays (80 serum samples against 36 viral strains) in approximately one month. We applied the sequencing-based assay to quantify the impact of influenza vaccination on neutralization titers against recent human H1N1 strains for individuals who had or had not also received a vaccine in the previous year. We found that the viral strain specificities of the neutralizing antibodies elicited by vaccination vary among individuals, and that vaccination induced a smaller increase in titers for individuals who had also received a vaccine the previous year-although the titers six months after vaccination were similar in individuals with and without the previous-year vaccination. We also identified a subset of individuals with low titers to a subclade of recent H1N1 even after vaccination. This study demonstrates the utility of high-throughput sequencing-based neutralization assays that enable titers to be simultaneously measured against many different viral strains. We provide a detailed experimental protocol (DOI: https://dx.doi.org/10.17504/protocols.io.kqdg3xdmpg25/v1) and a computational pipeline (https://github.com/jbloomlab/seqneut-pipeline) for the sequencing-based neutralization assays to facilitate the use of this method by others.
Collapse
Affiliation(s)
- Andrea N Loes
- Howard Hughes Medical Institute, Seattle, WA
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Rosario Araceli L Tarabi
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - John Huddleston
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Lisa Touyon
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Sook San Wong
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Samuel M S Cheng
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Nancy H L Leung
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - William W Hannon
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98109, USA
| | - Trevor Bedford
- Howard Hughes Medical Institute, Seattle, WA
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL
| | - Benjamin J Cowling
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Jesse D Bloom
- Howard Hughes Medical Institute, Seattle, WA
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
8
|
Petro-Turnquist E, Corder Kampfe B, Gadeken A, Pekarek MJ, Weaver EA. Multivalent Epigraph Hemagglutinin Vaccine Protects against Influenza B Virus in Mice. Pathogens 2024; 13:97. [PMID: 38392835 PMCID: PMC10892733 DOI: 10.3390/pathogens13020097] [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: 12/23/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Influenza B virus is a respiratory pathogen that contributes to seasonal epidemics, accounts for approximately 25% of global influenza infections, and can induce severe disease in young children. While vaccination is the most commonly used method of preventing influenza infections, current vaccines only induce strain-specific responses and have suboptimal efficacy when mismatched from circulating strains. Further, two influenza B virus lineages have been described, B/Yamagata-like and B/Victoria-like, and the limited cross-reactivity between the two lineages provides an additional barrier in developing a universal influenza B virus vaccine. Here, we report a novel multivalent vaccine using computationally designed Epigraph hemagglutinin proteins targeting both the B/Yamagata-like and B/Victoria-like lineages. When compared to the quadrivalent commercial vaccine, the Epigraph vaccine demonstrated increased breadth of neutralizing antibody and T cell responses. After lethal heterologous influenza B virus challenge, mice immunized with the Epigraph vaccine were completely protected against both weight loss and mortality. The superior cross-reactive immunity conferred by the Epigraph vaccine immunogens supports their continued investigation as a universal influenza B virus vaccine.
Collapse
Affiliation(s)
- Erika Petro-Turnquist
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Brigette Corder Kampfe
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
- Science Department, North Arkansas College, Harrison, AR 72601, USA
| | - Amber Gadeken
- College of Agricultural Sciences and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| | - Matthew J. Pekarek
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
9
|
Li M, Wang W, Chen J, Zhan Z, Xu M, Liu N, Ren L, You L, Zheng W, Shi H, Zhao Z, Huang C, Chen X, Zheng N, Lu W, Zhou X, Zhou J, Liao Q, Yang J, Jit M, Salje H, Yu H. Transplacental transfer efficiency of maternal antibodies against influenza A(H1N1)pdm09 virus and dynamics of naturally acquired antibodies in Chinese children: a longitudinal, paired mother-neonate cohort study. THE LANCET. MICROBE 2023; 4:e893-e902. [PMID: 37827184 DOI: 10.1016/s2666-5247(23)00181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND The 2009 pandemic H1N1 influenza A virus (A(H1N1)pdm09 virus) evolves rapidly and has continued to cause severe infections in children since its emergence in 2009. We aimed to characterise the kinetics of maternally and naturally acquired antibodies against historical A(H1N1)pdm09 strains and to assess the extent to which the response to heterologous strains following infection or vaccination affects observed A(H1N1)pdm09 strain-specific antibody titres in a Chinese paediatric population. METHODS In this retrospective study, we used residual serum samples from 528 mother-neonate pairs from a non-interventional, longitudinal cohort study in southern China conducted from Sept 20, 2013, to Aug 24, 2018, from six local hospitals in Anhua County, Hunan Province, China. Mother-neonate pairs were eligible for inclusion if the neonates were born after Sept 20, 2013, and their mothers had resided in the study sites for at least 3 months. We tested samples with a haemagglutination inhibition (HAI) assay to measure antibody levels against three historical A(H1N1)pdm09 strains that were antigenically similar to the strains that circulated during the 2009 pandemic (A/Hunan-Kaifu/SWL4204/2009 [SWL4204/09 strain], A/Hunan-Daxiang/SWL1277/2016 [SWL1277/16 strain], and A/Hunan-Yanfeng/SWL185/2018 [SWL185/18 strain]). We also determined the seroprevalence, geometric mean titres (GMTs), transfer ratio of maternal antibodies, and the dynamics of maternally and naturally acquired antibodies in children, from birth to 3 years of age. FINDINGS 1066 mother-neonate pairs were enrolled in the original cohort between Sept 20, 2013, and Oct 14, 2015. Of these, 528 pairs (523 mothers, 528 neonates) were selected for the present study. The median age of the mothers was 25 years (IQR 23 to 29). 291 (55%) of 528 children were boys and 237 (45%) were girls, and most children (452 [86%]) were breastfed before the age of 6 months. The GMTs and the seroprevalence for the SWL4204/09 strain were higher than those for the SWL1277/16 and SWL185/18 strains among mothers (GMTs: 10·4 [95% CI 9·8 to 11·1] vs 9·3 [8·7 to 9·8] vs 8·0 [7·5 to 8·4], p<0·0001; seroprevalence: 11·1% [95% CI 8·5 to 14·1] vs 6·9% [4·9 to 9·4] vs 4·6% [3·0 to 6·8], p=0·0003) and among neonates (GMTs: 10·7 [10·0 to 11·5] vs 9·4 [8·8 to 10·0] vs 8·1 [7·6 to 8·6], p<0·0001; seroprevalence: 13·4% [10·7 to 16·7] vs 8·7% [6·5 to 11·5] vs 6·1% [4·2 to 8·5], p=0·0002). Regardless of the A(H1N1)pdm09-specific strain, maternal antibodies could be transferred efficiently via the placenta (mean transfer ratios: 1·10 for SWL4204/09 vs 1·09 for SWL1277/16 vs 1·06 for SWL185/18; p=0·93). The A(H1N1)pdm09 strain-specific antibodies waned below the protective threshold of 1:40 within 2 months after birth. After maternal antibody waning, there were periodic increases and decreases in HAI antibody titres against three A(H1N1)pdm09 strains, and such increases were all significantly associated with a higher immune response to heterologous strains. Vaccination against the SWL4204/09 strain was associated with a poor response to the SWL185/18 strain (β-0·20, 95% CI -0·28 to -0·13; p<0·0001). INTERPRETATION Our findings suggest low pre-existing immunity against influenza A(H1N1)pdm09 virus among unvaccinated Chinese adult female and paediatric populations. This evidence, together with the rapid decay of maternal antibodies and the observed cross-reactivity among different A(H1N1)pdm09 strains, highlights the importance of accelerating maternal and paediatric influenza vaccination in China. FUNDING The Key Program of the National Natural Science Foundation of China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Mei Li
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Wei Wang
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Junbo Chen
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Zhifei Zhan
- Hunan Provincial Center for Disease Control and Prevention, Changsha, China
| | - Meng Xu
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Nuolan Liu
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Lingshuang Ren
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Lei You
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Wen Zheng
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Huilin Shi
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Zeyao Zhao
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Chaoyang Huang
- Hunan Provincial Center for Disease Control and Prevention, Changsha, China
| | - Xinhua Chen
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Nan Zheng
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Wanying Lu
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Xiaoyu Zhou
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Jiaxin Zhou
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Qiaohong Liao
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Juan Yang
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Hongjie Yu
- School of Public Health, Fudan University, Key Laboratory of Public Health Safety, Ministry of Education, Shanghai, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China; Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Cheung M, Chang C, Rathnasinghe R, Rossignol E, Zhang Y, Ferrari A, Patel H, Huang Y, Sanchez Guillen M, Scalzo T, Lee C, Otten GR, Settembre EC, Music N, Palladino G, Wen Y. Self-amplifying mRNA seasonal influenza vaccines elicit mouse neutralizing antibody and cell-mediated immunity and protect ferrets. NPJ Vaccines 2023; 8:150. [PMID: 37794051 PMCID: PMC10550923 DOI: 10.1038/s41541-023-00747-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023] Open
Abstract
Currently licensed influenza vaccines focus immune responses on viral hemagglutinin (HA), while the other major surface glycoprotein neuraminidase (NA) is not tightly controlled in inactivated vaccine formulations despite evidence that anti-NA antibodies reduce clinical disease. We utilized a bicistronic self-amplifying mRNA (sa-mRNA) platform encoding both HA and NA from four seasonal influenza strains, creating a quadrivalent influenza vaccine. sa-mRNA vaccines encoding an NA component induced the production of NA-inhibiting antibodies and CD4+ T-cell responses in both monovalent and quadrivalent formulations. Including NA in the vaccine enabled cross-neutralization against antigenically drifted strains and provided greater protection than HA alone upon A(H3N2) challenge in ferrets. These results demonstrate that next-generation bicistronic sa-mRNA vaccines expressing HA and NA induce potent antibodies against both viral coat proteins, as well as vaccine-specific cell-mediated immunity. When formulated as a quadrivalent seasonal influenza vaccine, the sa-mRNA platform provides an opportunity to increase the breadth of protection through cross-neutralizing anti-NA antibodies.
Collapse
Affiliation(s)
| | - Cheng Chang
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | | | - Yunfei Zhang
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | - Harsh Patel
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | - Yanjun Huang
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | - Tina Scalzo
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | - Changkeun Lee
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | | | - Nedzad Music
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA
| | | | - Yingxia Wen
- CSL Seqirus, 225 Wyman Street, Waltham, MA, 02451, USA.
| |
Collapse
|
11
|
Gao F, Liu X, Dang Y, Duan P, Xu W, Zhang X, Wang S, Luo J, Li X. AddaVax-Adjuvanted H5N8 Inactivated Vaccine Induces Robust Humoral Immune Response against Different Clades of H5 Viruses. Vaccines (Basel) 2022; 10:vaccines10101683. [PMID: 36298548 PMCID: PMC9612011 DOI: 10.3390/vaccines10101683] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 12/01/2022] Open
Abstract
Since some cases of human infections with H5N8 avian influenza virus have been reported and caused great concern in recent years, it is important to develop an effective vaccine for human use to prevent a potential H5N8 pandemic. In the present study, a vaccine candidate virus based on newly human-infected A/Astrakhan/3212/2020 H5N8 virus was constructed by reverse genetics (RG) technology. The immunogenicity of H5N8 whole virion inactivated vaccine was evaluated by various doses of vaccine antigen formulated with squalene-based adjuvant (AddaVax), aluminum hydroxide (Al(OH)3) or without adjuvant in mice. The results showed AddaVax-adjuvanted H5N8 inactivated vaccine could stimulate the mice to produce a stronger protective immune response with higher titers of IgG antibodies, hemagglutination inhibition (HI), neuraminidase inhibition (NI) and microneutralization (MN) antibodies than vaccine formulations with Al(OH)3 adjuvant or without adjuvant, and achieve a dose-sparing effect. Moreover, the AddaVax-adjuvanted formulation also exhibited potent cross-reactive response in HI antibodies against different clades of H5 viruses. A significant correlation and a curve fitting among HI, NI and MN were found by the correlation analysis to predict the protective effect of the vaccine. With these findings, our study demonstrates that AddaVax adjuvant can enhance the immunogenicity of H5N8 inactivated vaccine remarkably, and proposes an effective strategy for dealing with a potential H5N8 virus pandemic.
Collapse
|
12
|
Chang C, Music N, Cheung M, Rossignol E, Bedi S, Patel H, Safari M, Lee C, Otten GR, Settembre EC, Palladino G, Wen Y. Self-amplifying mRNA bicistronic influenza vaccines raise cross-reactive immune responses in mice and prevent infection in ferrets. Mol Ther Methods Clin Dev 2022; 27:195-205. [PMID: 36320414 PMCID: PMC9589142 DOI: 10.1016/j.omtm.2022.09.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022]
Abstract
Vaccines are the primary intervention against influenza. Currently licensed inactivated vaccines focus immunity on viral hemagglutinin (HA). Self-amplifying mRNA (sa-mRNA) vaccines offer an opportunity to generate immunity to multiple viral proteins, including additional neuraminidase (NA). This evaluation of a bicistronic approach for sa-mRNA vaccine development compared subgenomic promoter and internal ribosome entry site strategies and found consistent and balanced expression of both HA and NA proteins in transfected cells. In mice, sa-mRNA bicistronic A/H5N1 vaccines raised potent anti-HA and anti-NA neutralizing antibody responses and HA- or NA-specific CD4+ and CD8+ T cell responses. The addition of NA also boosted the cross-neutralizing response to heterologous A/H1N1. Similar immunogenicity results were obtained for bicistronic seasonal A/H3N2 and B/Yamagata vaccines. In ferrets, sa-mRNA bicistronic A/H1N1 vaccine fully protected lung from infection by homologous virus and showed significant reduction of viral load in upper respiratory tract, warranting further evaluation of sa-mRNA bicistronic vaccine in humans.
Collapse
Affiliation(s)
- Cheng Chang
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | - Nedzad Music
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | | | | | | | - Harsh Patel
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | | | | | | | | | | | - Yingxia Wen
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA,Corresponding author Yingxia Wen, CSL, 50 Hampshire Street, Cambridge, MA 02139, USA.
| |
Collapse
|
13
|
Ke W, Lai Q, Yang Y, Qian J, Ji X, He Z. Ratiometric Fluorescence Determination of Avian Influenza a Virus Subtype H1N1 DNA with Functionalized Quantum Dots and Gold Nanoparticles. ANAL LETT 2022. [DOI: 10.1080/00032719.2022.2052306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Wenmin Ke
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Qizhen Lai
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Yixia Yang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Jingjing Qian
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Xinghu Ji
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Zhike He
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Chanthavanich P, Versage E, Van Twuijver E, Hohenboken M. Antibody responses against heterologous A/H5N1 strains for an MF59-adjuvanted cell culture-derived A/H5N1 (aH5N1c) influenza vaccine in healthy pediatric subjects. Vaccine 2021; 39:6930-6935. [PMID: 34711436 DOI: 10.1016/j.vaccine.2021.10.010] [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: 04/13/2021] [Revised: 09/02/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Vaccines are the main prophylactic measure against pandemic influenza. Adjuvanted, cell culture-derived vaccines, which are not subject to limitations of egg-based vaccine production, have the potential to elicit an antibody response against heterologous strains and may be beneficial in the event of an A/H5N1 pandemic. METHODS A prespecified exploratory analysis of data from a phase 2, randomized, controlled, observer-blind multicenter trial (NCT01776554) to evaluate the immunogenicity of a MF59-adjuvanted, cell culture-based A/H5N1 influenza vaccine (aH5N1c), containing 7.5 µg hemagglutinin antigen per dose, in subjects 6 months through 17 years of age was conducted. Geometric mean titers (GMT) were determined using hemagglutination inhibition (HI) and microneutralization (MN) assays, and proportions of patients achieving seroconversion, HI and MN titers ≥ 1:40, and a 4-fold increase in MN titers against 5 heterologous strains (influenza A/H5N1 Anhui/2005, Egypt/2010, Hubei/2010, Indonesia/2005, and Vietnam/1203/2004) three weeks after administration of the second dose were assessed. RESULTS After the second dose, HI GMTs against heterologous strains increased between 8- and 40-fold, and MN GMTs increased 13- to 160-fold on Day 43 vs Day 1. On Day 43, 32-72% of subjects had HI titers ≥ 1:40 and achieved seroconversion against the heterologous strains. Using the MN assay, 84-100% of subjects had MN titers ≥ 1:40 and 83-100% achieved an at least 4-fold increase in MN titers against the heterologous strains. The highest responses were consistently against A/H5N1 Egypt/2010. CONCLUSIONS When given to children aged 6 months through 17 years, aH5N1c resulted in increased immunogenicity from baseline against all 5 heterologous A/H5N1 strains tested, demonstrating the potential of an MF59-adjuvanted, cell-derived A/H5N1 vaccine to provide cross-protection against other A/H5N1 strains (NCT01776554).
Collapse
Affiliation(s)
- Pornthep Chanthavanich
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Eve Versage
- Seqirus Inc., Clinical Development, Cambridge, USA
| | | | | |
Collapse
|
15
|
Lim J, Cheong Y, Kim YS, Chae W, Hwang BJ, Lee J, Jang YH, Roh YH, Seo SU, Seong BL. RNA-dependent assembly of chimeric antigen nanoparticles as an efficient H5N1 pre-pandemic vaccine platform. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102438. [PMID: 34256061 DOI: 10.1016/j.nano.2021.102438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 11/17/2022]
Abstract
Highly pathogenic avian influenza viruses (HPAIVs) pose a significant threat to human health, with high mortality rates, and require effective vaccines. We showed that, harnessed with novel RNA-mediated chaperone function, hemagglutinin (HA) of H5N1 HPAIV could be displayed as an immunologically relevant conformation on self-assembled chimeric nanoparticles (cNP). A tri-partite monomeric antigen was designed including: i) an RNA-interaction domain (RID) as a docking tag for RNA to enable chaperna function (chaperna: chaperone + RNA), ii) globular head domain (gd) of HA as a target antigen, and iii) ferritin as a scaffold for 24 mer-assembly. The immunization of mice with the nanoparticles (~46 nm) induced a 25-30 fold higher neutralizing capacity of the antibody and provided cross-protection from homologous and heterologous lethal challenges. This study suggests that cNP assembly is conducive to eliciting antibodies against the conserved region in HA, providing potent and broad protective efficacy.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Birds/virology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/therapeutic use
- Humans
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/chemistry
- Influenza Vaccines/immunology
- Influenza Vaccines/therapeutic use
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Mice
- Nanoparticles/chemistry
- Nanoparticles/therapeutic use
- Pandemics
- RNA/genetics
- RNA/immunology
- RNA/therapeutic use
Collapse
Affiliation(s)
- Jongkwan Lim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yucheol Cheong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young-Seok Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Wonil Chae
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Beom Jeung Hwang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jinhee Lee
- Department of Integrated OMICS for Biomedical Science, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yo Han Jang
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Biotechnology, Andong National University, Andong, Republic of Korea
| | - Young Hoon Roh
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Baik L Seong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Yonsei University, Seoul, Republic of Korea; Vaccine Innovative Technology Alliance-Korea, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|