1
|
Kim S, Flores RA, Moon SY, Lee SY, Altanzul B, Baek J, Choi EB, Lim H, Jang EY, Lee YK, Ouh IO, Kim WH. Design and Preliminary Immunogenicity Evaluation of Nipah Virus Glycoprotein G Epitope-Based Peptide Vaccine in Mice. Vaccines (Basel) 2025; 13:428. [PMID: 40333318 PMCID: PMC12031491 DOI: 10.3390/vaccines13040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
Background: The emergence of several paramyxoviruses, including Nipah virus (NiV), makes continued efforts in vaccine development as part of pandemic preparedness efforts necessary. Although NiV is a zoonotic pathogen with high case fatality, there is still no licensed vaccine. Methods: Herein, NiV attachment glycoprotein G (NiV-G), which is crucial to host cell receptor binding, was used to develop Nipah epitope-based peptide vaccines. A total of 39 B- and T-cell epitopes of NiV-G were shortlisted for peptide synthesis and evaluation using in silico analysis. Results: The in vitro antigenicity evaluation of the peptide candidates showed eight synthesized peptides (G7, stalk-domain epitopes) with relatively high binding to NiV-G antibody-positive serum (A450nm: 1.39-3.78). Moreover, nine-mer (9-mer) peptides were found to be less reactive than their longer peptide counterparts (15-30 aa, G7-1, and G7-4), but 9-mer activity was enhanced with cyclization (NPLPFREYK, A450nm: 2.66) and C-terminal amidation modification (NPLPFREYK-NH2, A450nm: 1.39). Subsequently, in vivo validation in immunized mice revealed the immunogenicity potential of the G7-1 peptide vaccine (30 aa, NENVNEKCKFTLPPLKIHECNISCPNPLPF) to elicit a strong antigen-specific antibody response against their homologous peptide antigen (I.V., A450nm: 1.48 ± 0.78; I.M., A450nm: 1.66 ± 0.66). However, antibody binding to recombinant NiV-G protein remained low, suggesting limited recognition to the native antigen. Conclusions: This study focused on the preliminary screening and validation of peptide vaccines using single formulations with minimal modifications in the peptide candidates. Our findings collectively show the immunogenic potential of the NiV-G stalk-based epitope peptide vaccine as a novel therapeutic for NiV and underscores the need for strategic design, delivery, and formulation optimization to enhance its protective efficacy and translational application.
Collapse
Affiliation(s)
- Seungyeon Kim
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Rochelle A. Flores
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| | - Seo Young Moon
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Seung Yun Lee
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| | - Bujinlkham Altanzul
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| | - Jiwon Baek
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Eun Bee Choi
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Heeji Lim
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Eun Young Jang
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Yoo-kyoung Lee
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - In-Ohk Ouh
- Division of Vaccine Development Coordination, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (S.K.); (S.Y.M.); (J.B.); (E.B.C.); (H.L.); (E.Y.J.); (Y.-k.L.)
| | - Woo H. Kim
- College of Veterinary Medicine & Institute of Animal Medicine, Gyeongsang National University, Jinju 52828, Gyeongsangnam-do, Republic of Korea; (R.A.F.); (S.Y.L.); (B.A.)
| |
Collapse
|
2
|
León AN, Rodriguez AJ, Richey ST, Torrents de la Pena A, Wolters RM, Jackson AM, Webb K, Creech CB, Yoder S, Mudd PA, Crowe JE, Han J, Ward AB. Structural mapping of polyclonal IgG responses to HA after influenza virus vaccination or infection. mBio 2025; 16:e0203024. [PMID: 39912630 PMCID: PMC11898601 DOI: 10.1128/mbio.02030-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
Cellular and molecular characterization of immune responses elicited by influenza virus infection and seasonal vaccination have informed efforts to improve vaccine efficacy, breadth, and longevity. Here, we use negative stain electron microscopy polyclonal epitope mapping (nsEMPEM) to structurally characterize the humoral IgG antibody responses to hemagglutinin (HA) from human patients vaccinated with a seasonal quadrivalent flu vaccine or infected with influenza A viruses. Our data show that both vaccinated and infected patients had humoral IgGs targeting highly conserved regions on both H1 and H3 subtype HAs, including the stem and anchor, which are targets for universal influenza vaccine design. Responses against H1 predominantly targeted the central stem epitope in infected patients and vaccinated donors, whereas head epitopes were more prominently targeted on H3. Responses against H3 were less abundant, but a greater diversity of H3 epitopes were targeted relative to H1. While our analysis is limited by sample size, on average, vaccinated donors responded to a greater diversity of epitopes on both H1 and H3 than infected patients. These data establish a baseline for assessing polyclonal antibody responses in vaccination and infection, providing a context for future vaccine trials and emphasizing the need for further characterization of protective responses toward conserved epitopes. (201 words)IMPORTANCESeasonal influenza viruses cause hundreds of thousands of deaths each year and up to a billion infections; under the proper circumstances, influenza A viruses with pandemic potential could threaten the lives of millions more. The variable efficacies of traditional influenza virus vaccines and the desire to prevent pandemic influenzas have motivated work toward finding a universal flu vaccine. Many promising universal flu vaccine candidates currently focus on guiding immune responses to highly conserved epitopes on the central stem of the influenza hemagglutinin viral fusion protein. To support the further development of these stem-targeting vaccine candidates, in this study, we use negative stain electron microscopy to assess the prevalence of central stem-targeting antibodies in individuals who were exposed to influenza antigens through traditional vaccination and/or natural infection during the 2018-2019 flu season.
Collapse
Affiliation(s)
- André Nicolás León
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California, USA
| | - Alesandra J. Rodriguez
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California, USA
| | - Sara T. Richey
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California, USA
| | - Alba Torrents de la Pena
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California, USA
| | - Rachael M. Wolters
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA
- Oregon Health & Science University, Portland, Oregon, USA
| | - Abigail M. Jackson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California, USA
| | - Katherine Webb
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - C. Buddy Creech
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Sandra Yoder
- Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Philip A. Mudd
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
- Department of Emergency Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - James E. Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Julianna Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California, USA
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, San Diego, California, USA
| |
Collapse
|
3
|
Chen C, Li M, Guo A, Guo P, Zhang W, Gu C, Wen G, Zhou H, Tao P. Addressing unexpected bacterial RNA safety concerns of E. coli produced influenza NP through CpG loaded mutant. NPJ Vaccines 2025; 10:32. [PMID: 39955275 PMCID: PMC11829966 DOI: 10.1038/s41541-025-01087-z] [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: 10/09/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
Influenza virus nucleoprotein (NP) is a promising target for universal influenza vaccines due to its conservation and high immunogenicity. Here, we uncovered a previously unknown factor that E. coli-produced NP carries bacterial RNA, which is crucial for its high immunogenicity but may pose safety and consistency concerns due to batch variability. To address these concerns, we developed a NP mutant (NPmut) that lacks RNA binding activity but can be loaded with CpG1826, a synthetic oligodeoxynucleotide adjuvant that has been used in the FDA-approved Hepatitis B vaccine. The CpG1826-loaded NPmut induced immune responses comparable to RNA-bound NP while eliminating safety risks. Additionally, the mixture of CpG1826-loaded NPmut and 3M2e protein (three tandem copies of the ectodomain of influenza M2 protein) provided enhanced protection against influenza viruses challenge. Our findings highlight the adjuvant activity of bacterial RNA in E. coli-produced NP and propose a safer strategy for developing universal influenza vaccines.
Collapse
Affiliation(s)
- Cen Chen
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Mengling Li
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Aili Guo
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Pengju Guo
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Wanpo Zhang
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Changqin Gu
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Guoyuan Wen
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, 430070, China
| | - Hongbo Zhou
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
- Hubei Hongshan Lab, Wuhan, Hubei, 430070, China.
| | - Pan Tao
- State Key Laboratory of Agricultural Microbiology, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
- Hubei Hongshan Lab, Wuhan, Hubei, 430070, China.
| |
Collapse
|
4
|
Mantus GE, Chopde AJ, Gorman J, Cominsky LY, Ourahmane A, Creanga A, Shimberg GD, Gillespie RA, Van Wazer DJ, Zhou T, Gajjala SR, Williams C, Maestle E, Reed DS, Serebryannyy L, Costner P, Holman L, Casazza JP, Koup RA, Dropulic LK, Kwong PD, McDermott AB, Kanekiyo M, Andrews SF. Vaccination with different group 2 influenza subtypes alters epitope targeting and breadth of hemagglutinin stem-specific human B cells. Sci Transl Med 2025; 17:eadr8373. [PMID: 39742506 DOI: 10.1126/scitranslmed.adr8373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
Abstract
The conserved influenza hemagglutinin stem, which is a target of cross-neutralizing antibodies, is now used in vaccine strategies focused on protecting against influenza pandemics. Antibody responses to group 1 stem have been extensively characterized, but little is known about group 2. Here, we characterized the stem-specific repertoire of individuals vaccinated with one of three group 2 influenza subtypes (H3, H7, or H10). Epitope mapping revealed two epitope supersites on the group 2 stem. Antibodies targeting the central epitope were broadly cross-reactive, whereas antibodies targeting the lower epitope had narrower breadth but higher potency against H3 subtypes. The ratio of B cells targeting each of the supersites varied with the vaccine subtype, leading to differences in the cross-reactivity of the B cell response. Our findings suggest that vaccine strategies targeting both group 2 stem epitopes would be complementary, eliciting broader and more potent protection against both seasonal and pandemic influenza strains.
Collapse
Affiliation(s)
- Grace E Mantus
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Ankita J Chopde
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Lauren Y Cominsky
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Amine Ourahmane
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Adrian Creanga
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Geoffrey D Shimberg
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Rebecca A Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - David J Van Wazer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Suprabhath R Gajjala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Connor Williams
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Emma Maestle
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Douglas S Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Leonid Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Pamela Costner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Lasonji Holman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Joseph P Casazza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Lesia K Dropulic
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Sarah F Andrews
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| |
Collapse
|
5
|
do Nascimento GM, de Oliveira PSB, Butt SL, Diel DG. Immunogenicity of chimeric hemagglutinins delivered by an orf virus vector platform against swine influenza virus. Front Immunol 2024; 15:1322879. [PMID: 38482020 PMCID: PMC10933025 DOI: 10.3389/fimmu.2024.1322879] [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: 10/17/2023] [Accepted: 01/22/2024] [Indexed: 04/05/2024] Open
Abstract
Orf virus (ORFV) is a large DNA virus that can harbor and efficiently deliver viral antigens in swine. Here we used ORFV as a vector platform to deliver chimeric hemagglutinins (HA) of Influenza A virus of swine (IAV-S). Vaccine development against IAV-S faces limitations posed by strain-specific immunity and the antigenic diversity of the IAV-S strains circulating in the field. A promising alternative aiming at re-directing immune responses on conserved epitopes of the stalk segment of the hemagglutinin (HA2) has recently emerged. Sequential immunization with chimeric HAs comprising the same stalk but distinct exotic head domains can potentially induce cross-reactive immune responses against conserved epitopes of the HA2 while breaking the immunodominance of the head domain (HA1). Here, we generated two recombinant ORFVs expressing chimeric HAs encoding the stalk region of a contemporary H1N1 IAV-S strain and exotic heads derived from either H6 or H8 subtypes, ORFVΔ121cH6/1 and ORFVΔ121cH8/1, respectively. The resulting recombinant viruses were able to express the heterologous protein in vitro. Further, the immunogenicity and cross-protection of these vaccine candidates were assessed in swine after sequential intramuscular immunization with OV-cH6/1 and OV-cH8/1, and subsequent challenge with divergent IAV-S strains. Humoral responses showed that vaccinated piglets presented increasing IgG responses in sera. Additionally, cross-reactive IgG and IgA antibody responses elicited by immunization were detected in sera and bronchoalveolar lavage (BAL), respectively, by ELISA against different viral clades and a diverse range of contemporary H1N1 IAV-S strains, indicating induction of humoral and mucosal immunity in vaccinated animals. Importantly, viral shedding was reduced in nasal swabs from vaccinated piglets after intranasal challenge with either Oh07 (gamma clade) or Ca09 (npdm clade) IAV-S strains. These results demonstrated the efficiency of ORFV-based vectors in delivering chimeric IAV-S HA-based vaccine candidates and underline the potential use of chimeric-HAs for prevention and control of influenza in swine.
Collapse
Affiliation(s)
- Gabriela Mansano do Nascimento
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Pablo Sebastian Britto de Oliveira
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
- Programa de Pós-graduação em Medicina Veterinária, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Salman Latif Butt
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Diego G. Diel
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
6
|
Mortazavi M, Pirbonyeh N, Javanmardi F, Emami A. Bioinformatics and Structural Analysis of Antigenic Variation in the Hemagglutinin Gene of the Influenza A(H1N1)pdm09 Virus Circulating in Shiraz (2013 to 2015). Microbiol Spectr 2023; 11:e0463022. [PMID: 37436149 PMCID: PMC10433955 DOI: 10.1128/spectrum.04630-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
Circulating influenza A virus provided an excellent opportunity to study the adaptation of the influenza A(H1N1)pdm09 virus to the human host. Particularly, due to the availability of sequences taken from isolates, we could monitor amino acid changes and the stability of mutations that occurred in hemagglutinin (HA). HA is crucial to viral infection because it binds to ciliated cell receptors and mediates the fusion of cells and viral membranes; because antibodies that bind to HA may block virus entry to the cell, this protein is subjected to high selective pressure. In this study, the locations of mutations in the structures of mutant HA were analyzed and the three-dimensional (3D) structures of these mutations were modeled in I-TASSER. Also, the location of these mutations was visualized and studied using Swiss PDB Viewer software and the PyMOL Molecular Graphics System. The crystal structure of the HA from A/California/07/2009 (3LZG) was used for further analysis. The new noncovalent bond formations in mutant luciferases were analyzed via WHAT IF and PIC, and protein stability was evaluated in the iStable server. We identified 33 and 23 mutations in A/Shiraz/106/2015 and A/California/07/2009 isolates, respectively; some mutations are located on the antigenic sites of Sa, Sb, Ca1, Ca2, and Cb HA1 and the fusion peptide of HA2. The results show that with the mutation some interactions are lost and new interactions are formed with other amino acids. The results of the free-energy analysis suggested that these new interactions have a destabilizing effect, which needs confirmation experimentally. IMPORTANCE Due to the fact that the mutations that occurred in the influenza virus HA cause the instability of the protein produced by the virus and antigenic changes and the escape of the virus from the immune system, the mutations that occurred in A/Shiraz/1/2013 were investigated in terms of energy level and stability. The mutations located in a globular portion of the HA are S188T, Q191H, S270P, K285Q, and P299L. On the other hand, the E374K, E46K-B, S124N-B, and I321V mutations are located in the stem portion of the HA (HA2). The change V252L mutation eliminates interactions with Ala181, Phe147, Leu151, and Trp153 and forms new interactions with Gly195, Asn264, Phe161, Met244, Tyr246, Leu165, and Trp167 which can change the stability of the HA structure. The K166Q mutation, which is located within the antigenic site Sa, causes the virus to escape from the immune response.
Collapse
Affiliation(s)
- Mojtaba Mortazavi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Neda Pirbonyeh
- Microbiology Department, Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Javanmardi
- Biostatistics Department, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Emami
- Microbiology Department, Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Andrews SF, Cominsky LY, Shimberg GD, Gillespie RA, Gorman J, Raab JE, Brand J, Creanga A, Gajjala SR, Narpala S, Cheung CSF, Harris DR, Zhou T, Gordon I, Holman L, Mendoza F, Houser KV, Chen GL, Mascola JR, Graham BS, Kwong PD, Widge A, Dropulic LK, Ledgerwood JE, Kanekiyo M, McDermott AB. An influenza H1 hemagglutinin stem-only immunogen elicits a broadly cross-reactive B cell response in humans. Sci Transl Med 2023; 15:eade4976. [PMID: 37075126 DOI: 10.1126/scitranslmed.ade4976] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Current yearly seasonal influenza vaccines primarily induce an antibody response directed against the immunodominant but continually diversifying hemagglutinin (HA) head region. These antibody responses provide protection against the vaccinating strain but little cross-protection against other influenza strains or subtypes. To focus the immune response on subdominant but more conserved epitopes on the HA stem that might protect against a broad range of influenza strains, we developed a stabilized H1 stem immunogen lacking the immunodominant head displayed on a ferritin nanoparticle (H1ssF). Here, we evaluated the B cell response to H1ssF in healthy adults ages 18 to 70 in a phase 1 clinical trial (NCT03814720). We observed both a strong plasmablast response and sustained elicitation of cross-reactive HA stem-specific memory B cells after vaccination with H1ssF in individuals of all ages. The B cell response was focused on two conserved epitopes on the H1 stem, with a highly restricted immunoglobulin repertoire unique to each epitope. On average, two-thirds of the B cell and serological antibody response recognized a central epitope on the H1 stem and exhibited broad neutralization across group 1 influenza virus subtypes. The remaining third recognized an epitope near the viral membrane anchor and was largely limited to H1 strains. Together, we demonstrate that an H1 HA immunogen lacking the immunodominant HA head produces a robust and broadly neutralizing HA stem-directed B cell response.
Collapse
Affiliation(s)
- Sarah F Andrews
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Lauren Y Cominsky
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Geoffrey D Shimberg
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Rebecca A Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Julie E Raab
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Joshua Brand
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Adrian Creanga
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Suprabhath R Gajjala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Crystal S F Cheung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Darcy R Harris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Ingelise Gordon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - LaSonji Holman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Floreliz Mendoza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Katherine V Houser
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Grace L Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Alicia Widge
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Lesia K Dropulic
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20902, USA
| |
Collapse
|
8
|
do Nascimento GM, Bugybayeva D, Patil V, Schrock J, Yadagiri G, Renukaradhya GJ, Diel DG. An Orf-Virus (ORFV)-Based Vector Expressing a Consensus H1 Hemagglutinin Provides Protection against Diverse Swine Influenza Viruses. Viruses 2023; 15:994. [PMID: 37112974 PMCID: PMC10147081 DOI: 10.3390/v15040994] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Influenza A viruses (IAV-S) belonging to the H1 subtype are endemic in swine worldwide. Antigenic drift and antigenic shift lead to a substantial antigenic diversity in circulating IAV-S strains. As a result, the most commonly used vaccines based on whole inactivated viruses (WIVs) provide low protection against divergent H1 strains due to the mismatch between the vaccine virus strain and the circulating one. Here, a consensus coding sequence of the full-length of HA from H1 subtype was generated in silico after alignment of the sequences from IAV-S isolates obtained from public databases and was delivered to pigs using the Orf virus (ORFV) vector platform. The immunogenicity and protective efficacy of the resulting ORFVΔ121conH1 recombinant virus were evaluated against divergent IAV-S strains in piglets. Virus shedding after intranasal/intratracheal challenge with two IAV-S strains was assessed by real-time RT-PCR and virus titration. Viral genome copies and infectious virus load were reduced in nasal secretions of immunized animals. Flow cytometry analysis showed that the frequency of T helper/memory cells, as well as cytotoxic T lymphocytes (CTLs), were significantly higher in the peripheral blood mononuclear cells (PBMCs) of the vaccinated groups compared to unvaccinated animals when they were challenged with a pandemic strain of IAV H1N1 (CA/09). Interestingly, the percentage of T cells was higher in the bronchoalveolar lavage of vaccinated animals in relation to unvaccinated animals in the groups challenged with a H1N1 from the gamma clade (OH/07). In summary, delivery of the consensus HA from the H1 IAV-S subtype by the parapoxvirus ORFV vector decreased shedding of infectious virus and viral load of IAV-S in nasal secretions and induced cellular protective immunity against divergent influenza viruses in swine.
Collapse
Affiliation(s)
- Gabriela Mansano do Nascimento
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA
| | - Dina Bugybayeva
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Veerupaxagouda Patil
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Jennifer Schrock
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Ganesh Yadagiri
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Gourapura J. Renukaradhya
- Department of Animal Sciences, Center for Food Animal Health, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - Diego G. Diel
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
9
|
Aartse A, Mortier D, Mooij P, Hofman S, van Haaren MM, Corcoran M, Karlsson Hedestam GB, Eggink D, Claireaux M, Bogers WMJM, van Gils MJ, Koopman G. Primary antibody response after influenza virus infection is first dominated by low-mutated HA-stem antibodies followed by higher-mutated HA-head antibodies. Front Immunol 2022; 13:1026951. [PMID: 36405682 PMCID: PMC9670313 DOI: 10.3389/fimmu.2022.1026951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/13/2022] [Indexed: 09/12/2023] Open
Abstract
Several studies have shown that the first encounter with influenza virus shapes the immune response to future infections or vaccinations. However, a detailed analysis of the primary antibody response is lacking as this is difficult to study in humans. It is therefore not known what the frequency and dynamics of the strain-specific hemagglutinin (HA) head- and stem-directed antibody responses are directly after primary influenza virus infection. Here, sera of twelve H1N1pdm2009 influenza virus-infected cynomolgus macaques were evaluated for HA-head and HA-stem domain antibody responses. We observed an early induction of HA-stem antibody responses, which was already decreased by day 56. In contrast, responses against the HA-head domain were low early after infection and increased at later timepoint. The HA-specific B cell repertoires in each animal showed diverse VH-gene usage with preferred VH-gene and JH-gene family usage for HA-head or HA-stem B cells but a highly diverse allelic variation within the VH-usage. HA-head B cells had shorter CDRH3s and higher VH-gene somatic hyper mutation levels relative to HA-stem B cells. In conclusion, our data suggest that HA-stem antibodies are the first to react to the infection while HA-head antibodies show a delayed response, but a greater propensity to enter the germinal center and undergo affinity maturation.
Collapse
Affiliation(s)
- Aafke Aartse
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, Netherlands
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, Amsterdam, Netherlands
| | - Daniella Mortier
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Petra Mooij
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Sam Hofman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Marlies M. van Haaren
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, Netherlands
| | - Martin Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet (KI), Stockholm, Sweden
| | | | - Dirk Eggink
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, Netherlands
| | - Mathieu Claireaux
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, Netherlands
| | | | - Marit J. van Gils
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, Netherlands
| | - Gerrit Koopman
- Department of Virology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| |
Collapse
|
10
|
Goll JB, Jain A, Jensen TL, Assis R, Nakajima R, Jasinskas A, Coughlan L, Cherikh SR, Gelber CE, Khan S, Huw Davies D, Meade P, Stadlbauer D, Strohmeier S, Krammer F, Chen WH, Felgner PL. The antibody landscapes following AS03 and MF59 adjuvanted H5N1 vaccination. NPJ Vaccines 2022; 7:103. [PMID: 36042229 PMCID: PMC9427073 DOI: 10.1038/s41541-022-00524-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 07/26/2022] [Indexed: 11/23/2022] Open
Abstract
Current seasonal and pre-pandemic influenza vaccines induce short-lived predominantly strain-specific and limited heterosubtypic responses. To better understand how vaccine adjuvants AS03 and MF59 may provide improved antibody responses to vaccination, we interrogated serum from subjects who received 2 doses of inactivated monovalent influenza A/Indonesia/05/2005 vaccine with or without AS03 or MF59 using hemagglutinin (HA) microarrays (NCT01317758 and NCT01317745). The arrays were designed to reflect both full-length and globular head HA derived from 17 influenza A subtypes (H1 to H16 and H18) and influenza B strains. We observed significantly increased strain-specific and broad homo- and heterosubtypic antibody responses with both AS03 and MF59 adjuvanted vaccination with AS03 achieving a higher titer and breadth of IgG responses relative to MF59. The adjuvanted vaccine was also associated with the elicitation of stalk-directed antibody. We established good correlation of the array antibody responses to H5 antigens with standard HA inhibition and microneutralization titers.
Collapse
Affiliation(s)
| | - Aarti Jain
- Vaccine R&D Center, Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA, USA
| | | | - Rafael Assis
- Vaccine R&D Center, Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA, USA
| | - Rie Nakajima
- Vaccine R&D Center, Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA, USA
| | - Algis Jasinskas
- Vaccine R&D Center, Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA, USA
| | - Lynda Coughlan
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - S Khan
- Vaccine R&D Center, Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA, USA
| | - D Huw Davies
- Vaccine R&D Center, Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA, USA
| | - Philip Meade
- Department of Microbiology, Icahn School of Medicine at Mount. Sinai, New York City, NY, USA
| | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount. Sinai, New York City, NY, USA
- Moderna Inc., Cambridge, MA, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount. Sinai, New York City, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount. Sinai, New York City, NY, USA
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Philip L Felgner
- Vaccine R&D Center, Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
11
|
Folschweiller N, Vanden Abeele C, Chu L, Van Damme P, García-Sastre A, Krammer F, Nachbagauer R, Palese P, Solórzano A, Bi D, David MP, Friel D, Innis BL, Koch J, Mallett CP, Rouxel RN, Salaun B, Vantomme V, Verheust C, Struyf F. Reactogenicity, safety, and immunogenicity of chimeric haemagglutinin influenza split-virion vaccines, adjuvanted with AS01 or AS03 or non-adjuvanted: a phase 1–2 randomised controlled trial. THE LANCET INFECTIOUS DISEASES 2022; 22:1062-1075. [DOI: 10.1016/s1473-3099(22)00024-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/26/2021] [Accepted: 01/06/2022] [Indexed: 12/19/2022]
|
12
|
Yegorov S, Celeste DB, Gomes KB, Ang JC, Vandenhof C, Wang J, Rybkina K, Tsui V, Stacey HD, Loeb M, Miller MS. Inactivated and live-attenuated seasonal influenza vaccines boost broadly neutralizing antibodies in children. Cell Rep Med 2022; 3:100509. [PMID: 35243417 PMCID: PMC8861809 DOI: 10.1016/j.xcrm.2022.100509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/16/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
The induction of broadly neutralizing antibodies (bNAbs) that target the hemagglutinin stalk domain is a promising strategy for the development of “universal” influenza virus vaccines. bNAbs can be boosted in adults by sequential exposure to heterosubtypic viruses through natural infection or vaccination. However, little is known about if or how bNAbs are induced by vaccination in more immunologically naive children. Here, we describe the impact of repeated seasonal influenza vaccination and vaccine type on induction of bNAbs against group 1 influenza viruses in a pediatric cohort enrolled in randomized controlled trials of seasonal influenza vaccination. Repeated seasonal vaccination results in significant boosting of a durable bNAb response. Boosting of serological bNAb titers is comparable within inactivated and live attenuated (LAIV) vaccinees and declines with age. These data provide insights into vaccine-elicited bNAb induction in children, which have important implications for the design of universal influenza vaccine modalities in this critical population. Repeated inactivated influenza vaccination boosts bNAbs Inactivated and live attenuated vaccines are similarly efficient at boosting bNAbs The magnitude of IIV and LAIV vaccine-elicited bNAb boosting declines with age
Collapse
Affiliation(s)
- Sergey Yegorov
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Daniel B. Celeste
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Kimberly Braz Gomes
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Jann C. Ang
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Colin Vandenhof
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Joanne Wang
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Ksenia Rybkina
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Vanessa Tsui
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Hannah D. Stacey
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Mark Loeb
- Michael G. DeGroote Institute for Infectious Disease Research, Health Research Methodology, Evidence, and Impact, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew S. Miller
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Corresponding author
| |
Collapse
|
13
|
Embong AK, Nguyen-Contant P, Wang J, Kanagaiah P, Chaves FA, Fitzgerald TF, Zhou Q, Kosoy G, Branche AR, Miller BL, Zand MS, Sangster MY, Topham DJ. Formation and Expansion of Memory B Cells against Coronavirus in Acutely Infected COVID-19 Individuals. Pathogens 2022; 11:186. [PMID: 35215130 PMCID: PMC8876169 DOI: 10.3390/pathogens11020186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
Infection with the β-coronavirus SARS-CoV-2 typically generates strong virus-specific antibody production. Antibody responses against novel features of SARS-CoV-2 proteins require naïve B cell activation, but there is a growing appreciation that conserved regions are recognized by pre-existing memory B cells (MBCs) generated by endemic coronaviruses. The current study investigated the role of pre-existing cross-reactive coronavirus memory in the antibody response to the viral spike (S) and nucleocapsid (N) proteins following SARS-CoV-2 infection. The breadth of reactivity of circulating antibodies, plasmablasts, and MBCs was analyzed. Acutely infected subjects generated strong IgG responses to the S protein, including the novel receptor binding domain, the conserved S2 region, and to the N protein. The response included reactivity to the S of endemic β-coronaviruses and, interestingly, to the N of an endemic α-coronavirus. Both mild and severe infection expanded IgG MBC populations reactive to the S of SARS-CoV-2 and endemic β-coronaviruses. Avidity of S-reactive IgG antibodies and MBCs increased after infection. Overall, findings indicate that the response to the S and N of SARS-CoV-2 involves pre-existing MBC activation and adaptation to novel features of the proteins, along with the potential of imprinting to shape the response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- A. Karim Embong
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA; (A.K.E.); (P.K.); (F.A.C.); (T.F.F.); (M.Y.S.)
| | | | - Jiong Wang
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14620, USA; (J.W.); (Q.Z.); (M.S.Z.)
| | - Preshetha Kanagaiah
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA; (A.K.E.); (P.K.); (F.A.C.); (T.F.F.); (M.Y.S.)
| | - Francisco A. Chaves
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA; (A.K.E.); (P.K.); (F.A.C.); (T.F.F.); (M.Y.S.)
| | - Theresa F. Fitzgerald
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA; (A.K.E.); (P.K.); (F.A.C.); (T.F.F.); (M.Y.S.)
| | - Qian Zhou
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14620, USA; (J.W.); (Q.Z.); (M.S.Z.)
| | - Gabrielle Kosoy
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY 14620, USA; (G.K.); (B.L.M.)
| | - Angela R. Branche
- Department of Medicine, University of Rochester Medical Center, Rochester, NY 14620, USA;
| | - Benjamin L. Miller
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY 14620, USA; (G.K.); (B.L.M.)
| | - Martin S. Zand
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14620, USA; (J.W.); (Q.Z.); (M.S.Z.)
| | - Mark Y. Sangster
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA; (A.K.E.); (P.K.); (F.A.C.); (T.F.F.); (M.Y.S.)
| | - David J. Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14620, USA; (A.K.E.); (P.K.); (F.A.C.); (T.F.F.); (M.Y.S.)
| |
Collapse
|
14
|
Caradonna TM, Schmidt AG. Protein engineering strategies for rational immunogen design. NPJ Vaccines 2021; 6:154. [PMID: 34921149 PMCID: PMC8683408 DOI: 10.1038/s41541-021-00417-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Antibody immunodominance refers to the preferential and asymmetric elicitation of antibodies against specific epitopes on a complex protein antigen. Traditional vaccination approaches for rapidly evolving pathogens have had limited success in part because of this phenomenon, as elicited antibodies preferentially target highly variable regions of antigens, and thus do not confer long lasting protection. While antibodies targeting functionally conserved epitopes have the potential to be broadly protective, they often make up a minority of the overall repertoire. Here, we discuss recent protein engineering strategies used to favorably alter patterns of immunodominance, and selectively focus antibody responses toward broadly protective epitopes in the pursuit of next-generation vaccines for rapidly evolving pathogens.
Collapse
Affiliation(s)
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
15
|
Rostaminia S, Aghaei SS, Farahmand B, Nazari R, Ghaemi A. Computational Design and Analysis of a Multi-epitope Against Influenza A virus. Int J Pept Res Ther 2021; 27:2625-2638. [PMID: 34539293 PMCID: PMC8435298 DOI: 10.1007/s10989-021-10278-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/28/2022]
Abstract
Influenza A viruses are among the most studied viruses, however no effective prevention against influenza infection has been developed. So, designing an effective vaccine against Influenza A virus is a critical issue in the field of medical biotechnology. For this reason, to combat this disease, we have designed a novel multi-epitope vaccine candidate based on the several conserved and potential linear B-cell and T-cell binding epitopes by using the in silico approach. This vaccine consists of an ER signal conserved sequence, the PADRE conserved epitope and two conserved epitopes of Influenza matrix protein 2. T-cell binding epitopes from Matrix protein 2 were predicted by in silico tools of epitope prediction. The selected epitopes were joined by flexible linkers and physicochemical properties, toxicity, and allergenecity were investigated. The designed vaccine was antigenic, immunogenic, and non-allergenic with suitable physicochemical properties and has higher solubility. The final multi-epitope construct was modeled, confirmed by different programs and the molecular interactions with immune receptors were considered. The molecular docking assay indicated the interactions with immune-stimulatory toll-like receptor 3 (TLR3) and major histocompatibility complex class I (MHCI). The HADDOCK and H DOCK servers were used to make docking analysis, respectively. The docking analysis indicated a strong and stable binding interaction between the vaccine construct with major histocompatibility complex (MHC) class I and toll-like receptor 3. Overall, the findings suggest that the current vaccine may be a promising vaccine to prevent Influenza infection.
Collapse
Affiliation(s)
- Samaneh Rostaminia
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | | | - Behrokh Farahmand
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, 69, P.O.Box: 1316943551, Tehran, Iran
| | - Raziye Nazari
- Department of Microbiology, Qom Branch, Islamic Azad University, Qom, Iran
| | - Amir Ghaemi
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, 69, P.O.Box: 1316943551, Tehran, Iran
| |
Collapse
|
16
|
Ferrara F, Del Rosario JMM, da Costa KAS, Kinsley R, Scott S, Fereidouni S, Thompson C, Kellam P, Gilbert S, Carnell G, Temperton N. Development of Lentiviral Vectors Pseudotyped With Influenza B Hemagglutinins: Application in Vaccine Immunogenicity, mAb Potency, and Sero-Surveillance Studies. Front Immunol 2021; 12:661379. [PMID: 34108964 PMCID: PMC8182064 DOI: 10.3389/fimmu.2021.661379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/05/2021] [Indexed: 12/16/2022] Open
Abstract
Influenza B viruses (IBV) cause respiratory disease epidemics in humans and are therefore components of seasonal influenza vaccines. Serological methods are employed to evaluate vaccine immunogenicity prior to licensure. However, classical methods to assess influenza vaccine immunogenicity such as the hemagglutination inhibition assay (HI) and the serial radial hemolysis assay (SRH), have been proven to have many limitations. As such, there is a need to develop innovative methods that can improve on these traditional assays and provide advantages such as ease of production and access, safety, reproducibility, and specificity. It has been previously demonstrated that the use of replication-defective viruses, such as lentiviral vectors pseudotyped with influenza A hemagglutinins in microneutralization assays (pMN) is a safe and sensitive alternative to study antibody responses elicited by natural influenza infection or vaccination. Consequently, we have produced Influenza B hemagglutinin-pseudotypes (IBV PV) using plasmid-directed transfection. To activate influenza B hemagglutinin, we have explored the use of proteases in increasing PV titers via their co-transfection during pseudotype virus production. When tested for their ability to transduce target cells, the influenza B pseudotypes produced exhibit tropism for different cell lines. The pseudotypes were evaluated as alternatives to live virus in microneutralization assays using reference sera standards, mouse and human sera collected during vaccine immunogenicity studies, surveillance sera from seals, and monoclonal antibodies (mAbs) against IBV. The influenza B pseudotype pMN was found to effectively detect neutralizing and cross-reactive responses in all assays and shows promise as an effective and versatile tool in influenza research.
Collapse
Affiliation(s)
- Francesca Ferrara
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent & University of Greenwich, Chatham, United Kingdom
| | - Joanne Marie M Del Rosario
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent & University of Greenwich, Chatham, United Kingdom.,Department of Physical Sciences & Mathematics, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| | - Kelly A S da Costa
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent & University of Greenwich, Chatham, United Kingdom
| | - Rebecca Kinsley
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent & University of Greenwich, Chatham, United Kingdom.,Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Simon Scott
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent & University of Greenwich, Chatham, United Kingdom
| | - Sasan Fereidouni
- Research Institute of Wildlife Ecology, Veterinary Medicine University, Vienna, Austria
| | - Craig Thompson
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Paul Kellam
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Sarah Gilbert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - George Carnell
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent & University of Greenwich, Chatham, United Kingdom.,Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent & University of Greenwich, Chatham, United Kingdom
| |
Collapse
|
17
|
Kerstetter LJ, Buckley S, Bliss CM, Coughlan L. Adenoviral Vectors as Vaccines for Emerging Avian Influenza Viruses. Front Immunol 2021; 11:607333. [PMID: 33633727 PMCID: PMC7901974 DOI: 10.3389/fimmu.2020.607333] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
It is evident that the emergence of infectious diseases, which have the potential for spillover from animal reservoirs, pose an ongoing threat to global health. Zoonotic transmission events have increased in frequency in recent decades due to changes in human behavior, including increased international travel, the wildlife trade, deforestation, and the intensification of farming practices to meet demand for meat consumption. Influenza A viruses (IAV) possess a number of features which make them a pandemic threat and a major concern for human health. Their segmented genome and error-prone process of replication can lead to the emergence of novel reassortant viruses, for which the human population are immunologically naïve. In addition, the ability for IAVs to infect aquatic birds and domestic animals, as well as humans, increases the likelihood for reassortment and the subsequent emergence of novel viruses. Sporadic spillover events in the past few decades have resulted in human infections with highly pathogenic avian influenza (HPAI) viruses, with high mortality. The application of conventional vaccine platforms used for the prevention of seasonal influenza viruses, such as inactivated influenza vaccines (IIVs) or live-attenuated influenza vaccines (LAIVs), in the development of vaccines for HPAI viruses is fraught with challenges. These issues are associated with manufacturing under enhanced biosafety containment, and difficulties in propagating HPAI viruses in embryonated eggs, due to their propensity for lethality in eggs. Overcoming manufacturing hurdles through the use of safer backbones, such as low pathogenicity avian influenza viruses (LPAI), can also be a challenge if incompatible with master strain viruses. Non-replicating adenoviral (Ad) vectors offer a number of advantages for the development of vaccines against HPAI viruses. Their genome is stable and permits the insertion of HPAI virus antigens (Ag), which are expressed in vivo following vaccination. Therefore, their manufacture does not require enhanced biosafety facilities or procedures and is egg-independent. Importantly, Ad vaccines have an exemplary safety and immunogenicity profile in numerous human clinical trials, and can be thermostabilized for stockpiling and pandemic preparedness. This review will discuss the status of Ad-based vaccines designed to protect against avian influenza viruses with pandemic potential.
Collapse
Affiliation(s)
- Lucas J. Kerstetter
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Stephen Buckley
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carly M. Bliss
- Division of Cancer & Genetics, Division of Infection & Immunity, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Nachbagauer R, Feser J, Naficy A, Bernstein DI, Guptill J, Walter EB, Berlanda-Scorza F, Stadlbauer D, Wilson PC, Aydillo T, Behzadi MA, Bhavsar D, Bliss C, Capuano C, Carreño JM, Chromikova V, Claeys C, Coughlan L, Freyn AW, Gast C, Javier A, Jiang K, Mariottini C, McMahon M, McNeal M, Solórzano A, Strohmeier S, Sun W, Van der Wielen M, Innis BL, García-Sastre A, Palese P, Krammer F. A chimeric hemagglutinin-based universal influenza virus vaccine approach induces broad and long-lasting immunity in a randomized, placebo-controlled phase I trial. Nat Med 2021; 27:106-114. [PMID: 33288923 DOI: 10.1038/s41591-020-1118-7] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/02/2020] [Indexed: 11/09/2022]
Abstract
Seasonal influenza viruses constantly change through antigenic drift and the emergence of pandemic influenza viruses through antigenic shift is unpredictable. Conventional influenza virus vaccines induce strain-specific neutralizing antibodies against the variable immunodominant globular head domain of the viral hemagglutinin protein. This necessitates frequent re-formulation of vaccines and handicaps pandemic preparedness. In this completed, observer-blind, randomized, placebo-controlled phase I trial (NCT03300050), safety and immunogenicity of chimeric hemagglutinin-based vaccines were tested in healthy, 18-39-year-old US adults. The study aimed to test the safety and ability of the vaccines to elicit broadly cross-reactive antibodies against the hemagglutinin stalk domain. Participants were enrolled into five groups to receive vaccinations with live-attenuated followed by AS03-adjuvanted inactivated vaccine (n = 20), live-attenuated followed by inactivated vaccine (n = 15), twice AS03-adjuvanted inactivated vaccine (n = 16) or placebo (n = 5, intranasal followed by intramuscular; n = 10, twice intramuscular) 3 months apart. Vaccination was found to be safe and induced a broad, strong, durable and functional immune response targeting the conserved, immunosubdominant stalk of the hemagglutinin. The results suggest that chimeric hemagglutinins have the potential to be developed as universal vaccines that protect broadly against influenza viruses.
Collapse
Affiliation(s)
- Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Moderna, Cambridge, MA, USA
| | - Jodi Feser
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Abdollah Naficy
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - David I Bernstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffrey Guptill
- Duke Early Phase Clinical Research Unit, Duke Clinical Research Institute, Durham, NC, USA
| | - Emmanuel B Walter
- Duke Early Phase Clinical Research Unit, Duke Clinical Research Institute, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrick C Wilson
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL, USA
- The Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohammad Amin Behzadi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Disha Bhavsar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carly Bliss
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christina Capuano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Veronika Chromikova
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carine Claeys
- GSK, Wavre, Belgium
- Spmt-Arista Asbl, Brussels, Belgium
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alec W Freyn
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Gast
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Andres Javier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaijun Jiang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Mariottini
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Monica McNeal
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alicia Solórzano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pfizer, Pearl River, NY, USA
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Bruce L Innis
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
19
|
Lei H, Gao T, Cen Q. Cross-protective immunity of the haemagglutinin stalk domain presented on the surface of Lactococcus lactis against divergent influenza viruses in mice. Virulence 2020; 12:12-19. [PMID: 33372841 PMCID: PMC7781637 DOI: 10.1080/21505594.2020.1857162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Most of the current approaches to influenza vaccine design focus on antibodies against influenza (HA). However, these influenza vaccines typically provide strain-specific protection against mostly homologous subtypes. There is an urgent need to develop a universal vaccine that confers cross-protection against influenza viruses. Of note, the HA stalk domain (HAsd) is a promising target for such an influenza vaccine. In this study, we generated recombinant Lactococcus lactis (L. lactis)/pNZ8150-phosphatidylglycerophosphate synthetase A (pgsA)-HAsd, in which pgsA was used as an anchor protein, and investigated the immunogenicity of HAsd in a mouse model by oral administration without the use of a mucosal adjuvant. Compared with L. lactis/pNZ8150-pgsA, mice were orally vaccinated with L. lactis/pNZ8150-pgsA-HAsd and then produced strong humoral and mucosal immune responses. Importantly, L. lactis/pNZ8150-pgsA-HAsd provided cross-protection against H5N1, H3N2 and H1N1 virus infections. Our data support the hypothesis that HAsd presented on the surface of L. lactis can provide cross-protective immunity against divergent influenza A viruses. Taken together, these findings suggest that L. lactis/pNZ8150-pgsA-HAsd can be considered an alternative approach to developing a novel universal vaccine during an influenza A pandemic. Abbreviations: HA, HAsd, HA stalk domain; L. lactis, Lactococcus lactis; SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis; IFA, immunofluorescence assay; PBS, phosphate-buffered saline; pgsA, phosphatidylglycerophosphate synthetase A; SPF, specific pathogen-free; CFU, colony-forming unit; BSL-3, biosafety level-3 laboratory; TCID50, 50% tissue culture infective dose; ELISA, enzyme-linked immunosorbent assay; OD, optical density; LTB, liable enterotoxin B subunit; CTB, cholera toxin B subunit.
Collapse
Affiliation(s)
- Han Lei
- College of Medicine, Southwest Jiaotong University , Chengdu, China
| | - Tong Gao
- College of Medicine, Southwest Jiaotong University , Chengdu, China
| | - Qianhong Cen
- College of Medicine, Southwest Jiaotong University , Chengdu, China
| |
Collapse
|
20
|
Carreño JM, McDonald JU, Hurst T, Rigsby P, Atkinson E, Charles L, Nachbagauer R, Behzadi MA, Strohmeier S, Coughlan L, Aydillo T, Brandenburg B, García-Sastre A, Kaszas K, Levine MZ, Manenti A, McDermott AB, Montomoli E, Muchene L, Narpala SR, Perera RAPM, Salisch NC, Valkenburg SA, Zhou F, Engelhardt OG, Krammer F. Development and Assessment of a Pooled Serum as Candidate Standard to Measure Influenza A Virus Group 1 Hemagglutinin Stalk-Reactive Antibodies. Vaccines (Basel) 2020; 8:vaccines8040666. [PMID: 33182279 PMCID: PMC7712758 DOI: 10.3390/vaccines8040666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/23/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022] Open
Abstract
The stalk domain of the hemagglutinin has been identified as a target for induction of protective antibody responses due to its high degree of conservation among numerous influenza subtypes and strains. However, current assays to measure stalk-based immunity are not standardized. Hence, harmonization of assay readouts would help to compare experiments conducted in different laboratories and increase confidence in results. Here, serum samples from healthy individuals (n = 110) were screened using a chimeric cH6/1 hemagglutinin enzyme-linked immunosorbent assay (ELISA) that measures stalk-reactive antibodies. We identified samples with moderate to high IgG anti-stalk antibody levels. Likewise, screening of the samples using the mini-hemagglutinin (HA) headless construct #4900 and analysis of the correlation between the two assays confirmed the presence and specificity of anti-stalk antibodies. Additionally, samples were characterized by a cH6/1N5 virus-based neutralization assay, an antibody-dependent cell-mediated cytotoxicity (ADCC) assay, and competition ELISAs, using the stalk-reactive monoclonal antibodies KB2 (mouse) and CR9114 (human). A “pooled serum” (PS) consisting of a mixture of selected serum samples was generated. The PS exhibited high levels of stalk-reactive antibodies, had a cH6/1N5-based neutralization titer of 320, and contained high levels of stalk-specific antibodies with ADCC activity. The PS, along with blinded samples of varying anti-stalk antibody titers, was distributed to multiple collaborators worldwide in a pilot collaborative study. The samples were subjected to different assays available in the different laboratories, to measure either binding or functional properties of the stalk-reactive antibodies contained in the serum. Results from binding and neutralization assays were analyzed to determine whether use of the PS as a standard could lead to better agreement between laboratories. The work presented here points the way towards the development of a serum standard for antibodies to the HA stalk domain of phylogenetic group 1.
Collapse
Affiliation(s)
- Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
| | - Jacqueline U. McDonald
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar EN6 3QG, UK; (J.U.M.); (T.H.); (L.C.)
| | - Tara Hurst
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar EN6 3QG, UK; (J.U.M.); (T.H.); (L.C.)
| | - Peter Rigsby
- Division of Analytical and Biological Sciences, National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar EN6 3QG, UK; (P.R.); (E.A.)
| | - Eleanor Atkinson
- Division of Analytical and Biological Sciences, National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar EN6 3QG, UK; (P.R.); (E.A.)
| | - Lethia Charles
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar EN6 3QG, UK; (J.U.M.); (T.H.); (L.C.)
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
| | - Mohammad Amin Behzadi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
- Global Health and Emerging Pathogens Institute, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA
| | - Boerries Brandenburg
- Janssen Vaccines & Prevention BV, 2333 CP Leiden, The Netherlands; (B.B.); (K.K.); (L.M.); (N.C.S.)
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
- Global Health and Emerging Pathogens Institute, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA
| | - Krisztian Kaszas
- Janssen Vaccines & Prevention BV, 2333 CP Leiden, The Netherlands; (B.B.); (K.K.); (L.M.); (N.C.S.)
| | - Min Z. Levine
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA;
| | | | - Adrian B. McDermott
- Vaccine Immunology Program (VIP), Vaccine Research Center (VRC), National Institutes of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA; (A.B.M.); (S.R.N.)
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy;
| | - Leacky Muchene
- Janssen Vaccines & Prevention BV, 2333 CP Leiden, The Netherlands; (B.B.); (K.K.); (L.M.); (N.C.S.)
| | - Sandeep R. Narpala
- Vaccine Immunology Program (VIP), Vaccine Research Center (VRC), National Institutes of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA; (A.B.M.); (S.R.N.)
| | - Ranawaka A. P. M. Perera
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (R.A.P.M.P.); (S.A.V.)
| | - Nadine C. Salisch
- Janssen Vaccines & Prevention BV, 2333 CP Leiden, The Netherlands; (B.B.); (K.K.); (L.M.); (N.C.S.)
| | - Sophie A. Valkenburg
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (R.A.P.M.P.); (S.A.V.)
| | - Fan Zhou
- Influenza Center, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway;
- K.G. Jebsen Center for influenza vaccines, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Othmar G. Engelhardt
- Division of Virology, National Institute for Biological Standards and Control (NIBSC), South Mimms, Potters Bar EN6 3QG, UK; (J.U.M.); (T.H.); (L.C.)
- Correspondence: (O.G.E.); (F.K.)
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA; (J.M.C.); (R.N.); (M.A.B.); (S.S.); (L.C.); (T.A.); (A.G.-S.)
- Correspondence: (O.G.E.); (F.K.)
| |
Collapse
|
21
|
Lopez CE, Legge KL. Influenza A Virus Vaccination: Immunity, Protection, and Recent Advances Toward A Universal Vaccine. Vaccines (Basel) 2020; 8:E434. [PMID: 32756443 PMCID: PMC7565301 DOI: 10.3390/vaccines8030434] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
Influenza virus infections represent a serious public health threat and account for significant morbidity and mortality worldwide due to seasonal epidemics and periodic pandemics. Despite being an important countermeasure to combat influenza virus and being highly efficacious when matched to circulating influenza viruses, current preventative strategies of vaccination against influenza virus often provide incomplete protection due the continuous antigenic drift/shift of circulating strains of influenza virus. Prevention and control of influenza virus infection with vaccines is dependent on the host immune response induced by vaccination and the various vaccine platforms induce different components of the local and systemic immune response. This review focuses on the immune basis of current (inactivated influenza vaccines (IIV) and live attenuated influenza vaccines (LAIV)) as well as novel vaccine platforms against influenza virus. Particular emphasis will be placed on how each platform induces cross-protection against heterologous influenza viruses, as well as how this immunity compares to and contrasts from the "gold standard" of immunity generated by natural influenza virus infection.
Collapse
Affiliation(s)
- Christopher E. Lopez
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin L. Legge
- Department of Microbiology and Immunology University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|