1
|
Li J, Liu M, Zhu J, Jiao Y, Zeng J. Ag-coated Au nanostar-based Lateral Flow Immunoassay for Highly Sensitive Influenza A virus antibody Detection in Colorimetric and Surface-Enhanced Raman Scattering (SERS) modes. Talanta 2025; 285:127351. [PMID: 39657522 DOI: 10.1016/j.talanta.2024.127351] [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: 09/23/2024] [Revised: 12/01/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Antibody testing for virus aids diagnosis, promotes vaccination and development, and evaluates antibody treatment efficacy. Hence, it is essential to examine and monitor antibody levels for accurate disease diagnosis and prevention. Lateral Flow Immunoassay (LFIA) is a technique that is known for its simplicity and speed, making it a popular choice for immediate detection. Noble metal nanoparticles are extensively employed in LFIA due to their exceptional colorimetric and Raman properties, which are a result of the LSPR effect. Au nanostars (Au NSs) have excellent SERS properties due to multiple sharp branches and more "hot spots" on the surface, while Ag nanoparticles (Ag NPs) have higher extinction coefficient and better refractive index sensitivity. The electromagnetic field strength on the surface of Ag-Au bimetallic nanomaterial is greatly enhanced, which further enhances the SERS intensity. In this work, we created a core-shell nanoparticle by combining Au NS as the core and Ag as the shell (Au NSMBA@Ag). We then inserted a Raman reporter molecule between the core and shell. Using this, we developed an LFIA platform that can detect influenza A virus antibodies in both colorimetric and Raman modes. The detection limit in colorimetric mode was 0.1 ng/mL, while in Raman mode it was 8.0 pg/mL, making it approximately 12 times more sensitive than the colorimetric mode. Furthermore, the method has shown excellent specificity, stability, and resistance to interference. Hence, this method can be applied to various fields such as environmental monitoring, clinical diagnosis, and food safety, showing great potential for future applications.
Collapse
Affiliation(s)
- Jingwen Li
- College of Chemistry and Chemical Engineering and State Key Laboratory of Heavy Oil Processing, China University of Petroleum (East China), Qingdao, 266580, China.
| | - Min Liu
- College of Chemistry and Chemical Engineering and State Key Laboratory of Heavy Oil Processing, China University of Petroleum (East China), Qingdao, 266580, China
| | - Jinyue Zhu
- College of Chemistry and Chemical Engineering and State Key Laboratory of Heavy Oil Processing, China University of Petroleum (East China), Qingdao, 266580, China
| | - Yunbing Jiao
- College of Chemistry and Chemical Engineering and State Key Laboratory of Heavy Oil Processing, China University of Petroleum (East China), Qingdao, 266580, China
| | - Jingbin Zeng
- College of Chemistry and Chemical Engineering and State Key Laboratory of Heavy Oil Processing, China University of Petroleum (East China), Qingdao, 266580, China.
| |
Collapse
|
2
|
Wu CY, Kao SE, Tseng YC, Hou JT, Wu LY, Chen JR. Pilot-scale production of a highly efficacious and stable monoglycosylated influenza split virus vaccine. Vaccine 2024; 42:2220-2228. [PMID: 38582606 DOI: 10.1016/j.vaccine.2023.11.038] [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: 07/14/2023] [Revised: 10/27/2023] [Accepted: 11/18/2023] [Indexed: 04/08/2024]
Abstract
The yearly epidemics and unpredictable outbreaks of influenza have raisedserious concernsglobally and led to prioritizing the development of an effective vaccine toprotectagainst newly emerging variants. Previously, we demonstrated that monoglycosylated influenza virus vaccines derived from A/California/7/2009 or an updated A/Brisbane/02/2018 (IVR-190) vaccine strain recommended by WHO are superior to fully glycosylated vaccines and could broadly protect against past and new coming H1N1 variants. However, whether such a monoglycosylated virus vaccine can be mass-produced to meet clinical demands and stable enough to provide consistent efficacy against H1N1 viruses remains unclear. Herein, we developed a platform for the pilot-scale production of the monoglycosylated split virus vaccine from the IVR-190 strain (IVR-190mg) with a robust and cost-effective manufacturing process. The critical parameters of inoculum dose, concentration of kifunensine, and optimized Endo H treatment process were comprehensively investigated. Several aims for preclinical studies of IVR-190mg were achieved, including [i] the execution of three engineering batch runs to validate lot-to-lot consistency, [ii] the establishment of IVR-190mg specifications to meet the acceptance criteria of a conventional influenza vaccine, [iii] an investigation of the stability profile of IVR-190mg, and completion of a safety evaluation by conducting an animal toxicology study. The toxicology study under GLP guidance found no systemic toxicity after rabbits were vaccinated with IVR-190mg. The serological data showed that IVR-190mg is highly immunogenic and effective in inducing a cross-strain protective level of antibody immune responses, including hemagglutination-inhibition titers, viral neutralization activity, and broad HA- and NA-inhibiting antibody titers against past and new H1N1 viruses. In conclusion, this study provides efficacy and safety profiles of IVR-190mg for further clinical study and shows that this vaccine without a glycan shield has great potential to be safe and protective against H1N1 variants.
Collapse
Affiliation(s)
- Chia-Ying Wu
- RuenHuei Biopharmaceuticals Inc., Taipei, Taiwan
| | - Shao-En Kao
- RuenHuei Biopharmaceuticals Inc., Taipei, Taiwan
| | | | - Jen-Tzu Hou
- RuenHuei Biopharmaceuticals Inc., Taipei, Taiwan
| | - Li-Yang Wu
- RuenHuei Biopharmaceuticals Inc., Taipei, Taiwan
| | | |
Collapse
|
3
|
Föhse K, Geckin B, Zoodsma M, Kilic G, Liu Z, Röring RJ, Overheul GJ, van de Maat J, Bulut O, Hoogerwerf JJ, Ten Oever J, Simonetti E, Schaal H, Adams O, Müller L, Ostermann PN, van de Veerdonk FL, Joosten LAB, Haagmans BL, van Crevel R, van Rij RP, GeurtsvanKessel C, de Jonge MI, Li Y, Domínguez-Andrés J, Netea MG. The impact of BNT162b2 mRNA vaccine on adaptive and innate immune responses. Clin Immunol 2023; 255:109762. [PMID: 37673225 DOI: 10.1016/j.clim.2023.109762] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/04/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
The mRNA-based BNT162b2 protects against severe disease and mortality caused by SARS-CoV-2 via induction of specific antibody and T-cell responses. Much less is known about its broad effects on immune responses against other pathogens. Here, we investigated the adaptive immune responses induced by BNT162b2 vaccination against various SARS-CoV-2 variants and its effects on the responsiveness of immune cells upon stimulation with heterologous stimuli. BNT162b2 vaccination induced effective humoral and cellular immunity against SARS-CoV-2 that started to wane after six months. We also observed long-term transcriptional changes in immune cells after vaccination. Additionally, vaccination with BNT162b2 modulated innate immune responses as measured by inflammatory cytokine production after stimulation - higher IL-1/IL-6 release and decreased IFN-α production. Altogether, these data expand our knowledge regarding the overall immunological effects of this new class of vaccines and underline the need for additional studies to elucidate their effects on both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Konstantin Föhse
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Büsra Geckin
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martijn Zoodsma
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Gizem Kilic
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Zhaoli Liu
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Rutger J Röring
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gijs J Overheul
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Josephine van de Maat
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ozlem Bulut
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jacobien J Hoogerwerf
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jaap Ten Oever
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elles Simonetti
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Heiner Schaal
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Ortwin Adams
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Lisa Müller
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Philipp Niklas Ostermann
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Frank L van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bart L Haagmans
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Marien I de Jonge
- Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany; TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Rumfelt KE, Fitzsimmons WJ, Truscon R, Monto AS, Martin ET, Lauring AS. A rapid and flexible microneutralization assay for serological assessment of influenza viruses. Influenza Other Respir Viruses 2023; 17:e13141. [PMID: 37127782 PMCID: PMC10174083 DOI: 10.1111/irv.13141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/03/2023] Open
Abstract
BACKGROUND Serological responses from influenza vaccination or infection are typically measured by hemagglutinin inhibition (HAI) or microneutralization (MN). Both methods are limited in feasibility, standardization, and generalizability to recent strains. We developed a luciferase MN (LMN) assay that combines the advantages of the conventional MN assay with the ease of the HAI assay. METHODS Sera were obtained from the HIVE study, a Michigan household cohort. Reverse genetics was used to generate recombinant influenza viruses expressing the hemagglutinin and neuraminidase of test strains, all other viral proteins from an A/WSN/1933 backbone, and a NanoLuc reporter. Serum neutralization of luciferase-expressing targets was quantified as a reduction in light emission from infected cells. Neutralization titers were measured for cell- and egg-adapted versions of A/Hong Kong/4801/2014 and A/Singapore/INFIMH-16-0019/2016 and compared to HAI titers against egg-grown antigens. RESULTS Three hundred thirty-three sera were collected from 259 participants between May 2016 and July 2018. Sampled participants were 7-68 years of age, and >80% were vaccinated against influenza. HAI and LMN titers were correlated for A/Hong Kong/4801/2014 (ρ = 0.52, p ≤ 0.01) and A/Singapore/INFIMH-16-0019/2016 (ρ = 0.79, p ≤ 0.01). LMN titers were lower for cell strains compared to egg strains (A/Hong Kong/4801/2014 mean log2 fold change = -2.66, p ≤ 0.01 and A/Singapore/INFIMH-16-0019/2016 mean log2 fold change = -3.15, p ≤ 0.01). CONCLUSIONS The LMN assay was feasible using limited sample volumes and able to differentiate small antigenic differences between egg-adapted and cell-derived strains. The correspondence of these results with the commonly used HAI confirms the utility of this assay for high-throughput studies of correlates of protection and vaccine response.
Collapse
Affiliation(s)
- Kalee E. Rumfelt
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - William J. Fitzsimmons
- Division of Infectious Diseases, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Rachel Truscon
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Arnold S. Monto
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Emily T. Martin
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Adam S. Lauring
- Division of Infectious Diseases, Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
- Department of Microbiology and ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
5
|
Ren W, Pei S, Jiang W, Zhao M, Jiang L, Liu H, Yi Y, Hui M, Li J. A replication-deficient H9N2 influenza virus carrying H5 hemagglutinin conferred protection against H9N2 and H5N1 influenza viruses in mice. Front Microbiol 2022; 13:1042916. [PMID: 36458187 PMCID: PMC9705590 DOI: 10.3389/fmicb.2022.1042916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 10/26/2022] [Indexed: 05/07/2024] Open
Abstract
H5N1 and H9N2 influenza viruses have been reported to cause human infections and are believed to have pandemic potential. The vaccine is an effective tool to prevent influenza virus infection. However, inactivated influenza vaccines sometimes result in low antigenicity as result leads to generating of incomplete immune protection in the form of low cellular and humoral immunity. While the low temperature adapted, traditional live attenuated influenza vaccine (LAIV) is associated with the potential risk to revert to a virulent phenotype, there appears an essential need for an alternative potent methodology to design and develop influenza vaccines with substantial safety and efficacy which may confer solid protection against H9N2 or H5N1 influenza virus infections. In the present study, a replication-deficient recombinant influenza virus, WM01ma-HA(H5), expressing hemagglutinin (HA) of both H9N2 and H5N1 subtypes was developed. The chimeric gene segment expressing HA(H5), was designed using the sequence of an open reading frame (ORF) of HA adopted from A/wild duck/Hunan/021/2005(H5N1)(HN021ma) which was flanked by the NA packaging signals of mouse-adapted strain A/Mink/Shandong/WM01/2014(H9N2)(WM01ma). Due to the absence of ORF of structural protein NA, the replication of this engineered H9N2 influenza viruses WM01ma-HA(H5) was hampered in vitro and in vivo but was well competent in MDCK cells stably expressing the NA protein of WM01ma. Intranasal vaccination of mice with WM01ma-HA(H5) stimulated robust immune response without any clinical signs and conferred complete protection from infection by H5N1 or H9N2 subtype influenza viruses.
Collapse
Affiliation(s)
- Weigang Ren
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Shuli Pei
- Henan Vocational College of Agriculture, Zhongmu, China
| | - Wenming Jiang
- Laboratory of Surveillance for Avian Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Meixia Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Le Jiang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Honggang Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Yongxiang Yi
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
- The Clinical Infectious Disease Center of Nanjing, Nanjing, China
| | - Mizhou Hui
- School of Life Science, Northeast Agricultural University, Harbin, China
| | - Junwei Li
- Department of Infectious Diseases, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, China
- The Clinical Infectious Disease Center of Nanjing, Nanjing, China
| |
Collapse
|
6
|
Hu K, Palmieri E, Samnuan K, Ricchetti B, Oldrini D, McKay PF, Wu G, Thorne L, Fooks AR, McElhinney LM, Goharriz H, Golding M, Shattock RJ, Micoli F. Generalized Modules for Membrane Antigens (GMMA), an outer membrane vesicle-based vaccine platform, for efficient viral antigen delivery. J Extracell Vesicles 2022; 11:e12247. [PMID: 36377074 PMCID: PMC9663859 DOI: 10.1002/jev2.12247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/19/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
Vaccine platforms enable fast development, testing, and manufacture of more affordable vaccines. Here, we evaluated Generalized Modules for Membrane Antigens (GMMA), outer membrane vesicles (OMVs) generated by genetically modified Gram-negative bacteria, as a vaccine platform for viral pathogens. Influenza A virus hemagglutinin (HA), either physically mixed with GMMA (HA+STmGMMA mix), or covalently linked to GMMA surface (HA-STmGMMA conjugate), significantly increased antigen-specific humoral and cellular responses, with HA-STmGMMA conjugate inducing further enhancement than HA+STmGMMA mix. HA-STmGMMA conjugate protected mice from lethal challenge. The versatility for this platform was confirmed by conjugation of rabies glycoprotein (RABVG) onto GMMA through the same method. RABVG+STmGMMA mix and RABVG-STmGMMA conjugate exhibited similar humoral and cellular response patterns and protection efficacy as the HA formulations, indicating relatively consistent responses for different vaccines based on the GMMA platform. Comparing to soluble protein, GMMA was more efficiently taken up in vivo and exhibited a B-cell preferential uptake in the draining lymph nodes (LNs). Together, GMMA enhances immunity against viral antigens, and the platform works well with different antigens while retaining similar immunomodulatory patterns. The findings of our study imply the great potential of GMMA-based vaccine platform also against viral infectious diseases.
Collapse
Affiliation(s)
- Kai Hu
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Karnyart Samnuan
- Department of Infectious Diseases, Imperial College London, London, UK
| | | | - Davide Oldrini
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Paul F McKay
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Guanghui Wu
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Leigh Thorne
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Anthony R Fooks
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Lorraine M McElhinney
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Hooman Goharriz
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Megan Golding
- Animal and Plant Health Agency (APHA), OIE Rabies Reference Laboratory, New Haw, Addlestone, Surrey, UK
| | - Robin J Shattock
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
7
|
Zhao N, Grund C, Beer M, Wang G, Harder TC. Tetraplex Fluorescent Microbead-Based Immunoassay for the Serodiagnosis of Newcastle Disease Virus and Avian Influenza Viruses in Poultry Sera. Pathogens 2022; 11:pathogens11091059. [PMID: 36145491 PMCID: PMC9505202 DOI: 10.3390/pathogens11091059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/03/2022] Open
Abstract
Virulent Newcastle disease virus (NDV) as well as highly pathogenic avian influenza (HPAIV) subtypes H5 and H7 induce contagious and lethal systemic disease in poultry. In contrast, low pathogenic AIV H5 and H7 may circulate clinically unnoticed in poultry but eventually generate HPAIV. Low pathogenic NDV strains are widely used as live-attenuated vaccines against ND. Serological tools are essential to conduct active surveillance for infections with notifiable AIV-H5, -H7 and to control vaccination against NDV and HPAIV in poultry populations. Here, recombinant nucleocapsid proteins (NP) of AIV and NDV, and haemagglutinin protein fragment-1 (HA1) of AIV subtypes H5 and H7 were expressed in E. coli. Purification and refolding were required before coating fluorescent microspheres via streptavidin-biotin linkage. The tetraplexed inhibition fluorescent microsphere immunoassay (iFMIA) was then assembled for analysis on a Luminex®-like platform (Bioplex®) using murine monoclonal antibodies specific for each of the four targets. The assay was evaluated by testing galliform poultry sera derived from experimental infections (n = 257) and from farms (n = 250), respectively. The tetraplex iFMIA compared favorably with commercially available ELISAs and the “gold standard” hemagglutination inhibition assay. Tetraplexed iFMIA provided a specific and sensitive tool to detect and discriminate AIV- and NDV-specific antibodies in the sera of galliform poultry.
Collapse
Affiliation(s)
- Na Zhao
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Suedufer 10, 17493 Greifswald, Germany
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510030, China
| | - Christian Grund
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Suedufer 10, 17493 Greifswald, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Suedufer 10, 17493 Greifswald, Germany
| | - Gang Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou 510030, China
| | - Timm C. Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, Suedufer 10, 17493 Greifswald, Germany
- Correspondence: ; Tel.: +49-38351-7-15-46
| |
Collapse
|
8
|
Haveri A, Ikonen N, Savolainen-Kopra C, Julkunen I. Long-lasting heterologous antibody responses after sequential vaccination with A/Indonesia/5/2005 and A/Vietnam/1203/2004 pre-pandemic influenza A(H5N1) virus vaccines. Vaccine 2020; 39:402-411. [PMID: 33246672 DOI: 10.1016/j.vaccine.2020.11.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 11/04/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Avian influenza A(H5N1) viruses have caused sporadic infections in humans and thus they pose a significant global health threat. Among symptomatic patients the case fatality rate has been ca. 50%. H5N1 viruses exist in multiple clades and subclades and several candidate vaccines have been developed to prevent A(H5N1) infection as a principal measure for preventing the disease. METHODS Serum antibodies against various influenza A(H5N1) clade viruses were measured in adults by ELISA-based microneutralization and haemagglutination inhibition tests before and after vaccination with two different A(H5N1) vaccines in 2009 and 2011. RESULTS Two doses of AS03-adjuvanted A/Indonesia/5/2005 vaccine induced good homologous but poor heterologous neutralizing antibody responses against different clade viruses. However, non-adjuvanted A/Vietnam/1203/2004 booster vaccination in 2011 induced very strong and long-lasting homologous and heterologous antibody responses while homologous response remained weak in naïve subjects. CONCLUSIONS Sequential vaccination with two different A(H5N1) pre-pandemic vaccines induced long-lasting high level cross-clade immunity against influenza A(H5N1) strains, thus supporting a prime-boost vaccination strategy in pandemic preparedness plans.
Collapse
Affiliation(s)
- Anu Haveri
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland.
| | - Niina Ikonen
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland
| | - Carita Savolainen-Kopra
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland
| | - Ilkka Julkunen
- Expert Microbiology Unit, Department of Health Security, Finnish Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland; Institute of Biomedicine, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520 Turku, Finland
| |
Collapse
|
9
|
Wang Y, Lv Y, Niu X, Dong J, Feng P, Li Q, Xu W, Li J, Li C, Li J, Luo J, Li Z, Liu Y, Tan YJ, Pan W, Chen L. L226Q Mutation on Influenza H7N9 Virus Hemagglutinin Increases Receptor-Binding Avidity and Leads to Biased Antigenicity Evaluation. J Virol 2020; 94:e00667-20. [PMID: 32796071 PMCID: PMC7527056 DOI: 10.1128/jvi.00667-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/27/2020] [Indexed: 11/20/2022] Open
Abstract
Since the first outbreak in 2013, the influenza A (H7N9) virus has continued emerging and has caused over five epidemic waves. Suspected antigenic changes of the H7N9 virus based on hemagglutination inhibition (HI) assay during the fifth outbreak have prompted the update of H7N9 candidate vaccine viruses (CVVs). In this study, we comprehensively compared the serological cross-reactivities induced by the hemagglutinins (HAs) of the earlier CVV A/Anhui/1/2013 (H7/AH13) and the updated A/Guangdong/17SF003/2016 (H7/GD16). We found that although H7/GD16 showed poor HI cross-reactivity to immune sera from mice and rhesus macaques vaccinated with either H7/AH13 or H7/GD16, the cross-reactive neutralizing antibodies between H7/AH13 and H7/GD16 were comparably high. Passive transfer of H7/AH13 immune sera also provided complete protection against the lethal challenge of H7N9/GD16 virus in mice. Analysis of amino acid mutations in the HAs between H7/AH13 and H7/GD16 revealed that L226Q substitution increases the HA binding avidity to sialic acid receptors on red blood cells, leading to decreased HI titers against viruses containing HA Q226 and thus resulting in a biased antigenic evaluation based on HI assay. These results suggest that amino acids located in the receptor-binding site could mislead the evaluation of antigenic variation by solely impacting the receptor-binding avidity to red blood cells without genuine contribution to antigenic drift. Our study highlighted that viral receptor-binding avidity and combination of multiple serological assays should be taken into consideration in evaluating and selecting a candidate vaccine virus of H7N9 and other subtypes of influenza viruses.IMPORTANCE The HI assay is a standard method for profiling the antigenic characterization of influenza viruses. Suspected antigenic changes based on HI divergency in H7N9 viruses during the 2016-2017 wave prompted the recommendation of new H7N9 candidate vaccine viruses (CVVs). In this study, we found that the L226Q substitution in HA of A/Guangdong/17SF003/2016 (H7/GD16) increased the viral receptor-binding avidity to red blood cells with no impact on the antigenicity of H7N9 virus. Although immune sera raised by an earlier vaccine strain (H7/AH13) showed poor HI titers against H7/GD16, the H7/AH13 immune sera had potent cross-neutralizing antibody titers against H7/GD16 and could provide complete passive protection against H7N9/GD16 virus challenge in mice. Our study highlights that receptor-binding avidity might lead to biased antigenic evaluation by using the HI assay. Other serological assays, such as the microneutralization (MN) assay, should be considered a complementary indicator for analysis of antigenic variation and selection of influenza CVVs.
Collapse
Affiliation(s)
- Yang Wang
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunhua Lv
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuefeng Niu
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ji Dong
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pei Feng
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qinming Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Xu
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiashun Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chufang Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiahui Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jia Luo
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhixia Li
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yichu Liu
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yee-Joo Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore, Singapore
| | - Weiqi Pan
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling Chen
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Blakney AK, Zhu Y, McKay PF, Bouton C, Yeow J, Tang J, Hu K, Samnuan K, Grigsby CL, Shattock RJ, Stevens MM. Big Is Beautiful: Enhanced saRNA Delivery and Immunogenicity by a Higher Molecular Weight, Bioreducible, Cationic Polymer. ACS NANO 2020; 14:5711-5727. [PMID: 32267667 PMCID: PMC7304921 DOI: 10.1021/acsnano.0c00326] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/08/2020] [Indexed: 05/18/2023]
Abstract
Self-amplifying RNA (saRNA) vaccines are highly advantageous, as they result in enhanced protein expression compared to mRNA (mRNA), thus minimizing the required dose. However, previous delivery strategies were optimized for siRNA or mRNA and do not necessarily deliver saRNA efficiently due to structural differences of these RNAs, thus motivating the development of saRNA delivery platforms. Here, we engineer a bioreducible, linear, cationic polymer called "pABOL" for saRNA delivery and show that increasing its molecular weight enhances delivery both in vitro and in vivo. We demonstrate that pABOL enhances protein expression and cellular uptake via both intramuscular and intradermal injection compared to commercially available polymers in vivo and that intramuscular injection confers complete protection against influenza challenge. Due to the scalability of polymer synthesis and ease of formulation preparation, we anticipate that this polymer is highly clinically translatable as a delivery vehicle for saRNA for both vaccines and therapeutics.
Collapse
Affiliation(s)
- Anna K. Blakney
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Yunqing Zhu
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
- School
of Materials Science and Engineering, Tongji
University, Shanghai, 200092, China
| | - Paul F. McKay
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Clément
R. Bouton
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Jonathan Yeow
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
| | - Jiaqing Tang
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
| | - Kai Hu
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Karnyart Samnuan
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Christopher L. Grigsby
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 65, Sweden
| | - Robin J. Shattock
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 65, Sweden
| |
Collapse
|
11
|
Gauger PC, Vincent AL. Serum Virus Neutralization Assay for Detection and Quantitation of Serum Neutralizing Antibodies to Influenza A Virus in Swine. Methods Mol Biol 2020; 2123:321-333. [PMID: 32170698 DOI: 10.1007/978-1-0716-0346-8_23] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The serum virus neutralization (SVN) assay is a serological test used to detect the presence and magnitude of functional systemic antibodies that prevent infectivity of a virus. The SVN assay is a highly sensitive and specific test that may be applied to influenza A viruses (IAV) in swine to measure the titer of neutralizing antibodies postexposure, postvaccination, or after passive transfer of maternally derived antibody (MDA). Conventional SVN methods performed in vitro are based on inhibition of virus infectivity in cell culture in the presence of neutralizing antibodies in serum. Titer determination may be based on the presence or absence of cytopathic effect or evidence of viral infection using an immunoreactive technique. The SVN assay is relatively inexpensive using standard laboratory equipment, although it requires cell culture, more time and labor, and technical skill to conduct the assay compared to other serological methods. The SVN test may be used to evaluate the level of serological cross-reactivity between IAV exposure or vaccine antisera and heterologous influenza viruses that may correlate with cross-protection in the host.
Collapse
Affiliation(s)
- Phillip C Gauger
- Veterinary Diagnostic Laboratory, Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| | - Amy L Vincent
- Virus and Prion Research Unit, National Animal Disease Center, USDA, Agricultural Research Service, Ames, IA, USA
| |
Collapse
|
12
|
Corder BN, Bullard BL, DeBeauchamp JL, Ilyushina NA, Webby RJ, Weaver EA. Influenza H1 Mosaic Hemagglutinin Vaccine Induces Broad Immunity and Protection in Mice. Vaccines (Basel) 2019; 7:vaccines7040195. [PMID: 31771231 PMCID: PMC6963302 DOI: 10.3390/vaccines7040195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022] Open
Abstract
Annually, influenza A virus (IAV) infects ~5-10% of adults and 20-30% of children worldwide. The primary resource to protect against infection is by vaccination. However, vaccination only induces strain-specific and transient immunity. Vaccine strategies that induce cross-protective immunity against the broad diversity of IAV are needed. Here we developed and tested a novel mosaic H1 HA immunogen. The mosaic immunogen was optimized in silico to include the most potential B and T cell epitopes (PBTE) across a diverse population of human H1 IAV. Phylogenetic analysis showed that the mosaic HA localizes towards the non-pandemic 2009 strains which encompasses the broadest diversity in the H1 IAV population. We compared the mosaic H1 immunogen to wild-type HA immunogens and the commercial inactivated influenza vaccine, Fluzone. When analyzed by ELISA, the mosaic immunogen induced stronger antibody responses against all four diverse H1 HA proteins. When analyzing T cell responses, again the mosaic immunogen induced stronger cellular immunity against all 4 diverse HA strains. Not only was the magnitude of T cell responses strongest in mosaic immunized mice, the number of epitopes recognized was also greater. The mosaic vaccinated mice showed strong cross-protection against challenges with three divergent IAV strains. These data show that the mosaic immunogen induces strong cross-protective immunity and should be investigated further as a universal influenza vaccine.
Collapse
Affiliation(s)
- Brigette N. Corder
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
| | - Brianna L. Bullard
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
| | - Jennifer L. DeBeauchamp
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.L.D.); (R.J.W.)
| | - Natalia A. Ilyushina
- Division of Biotechnology Review and Research II, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA;
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.L.D.); (R.J.W.)
| | - Eric A. Weaver
- School of Biological Sciences, Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (B.N.C.); (B.L.B.)
- Correspondence:
| |
Collapse
|
13
|
Tambyah PA, Oon J, Asli R, Kristanto W, Hwa SH, Vang F, Karwal L, Fuchs J, Santangelo JD, Gordon GS, Thomson C, Rao R, Dean H, Das SC, Stinchcomb DT. An inactivated enterovirus 71 vaccine is safe and immunogenic in healthy adults: A phase I, double blind, randomized, placebo-controlled, study of two dosages. Vaccine 2019; 37:4344-4353. [PMID: 31230881 DOI: 10.1016/j.vaccine.2019.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/10/2019] [Accepted: 06/12/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Hand, foot and mouth disease (HFMD), especially that caused by enterovirus 71 (EV71) infection, is a public health concern in the Asia-Pacific region. We report a phase I clinical trial of an EV71 candidate vaccine (INV21) based on a binary ethylenimine inactivated B2 sub-genotype formulated with aluminum hydroxide. METHODS In this double-blind, placebo-controlled, randomized, dose escalation study adult volunteers received two vaccinations 28 days apart of low or high dose formulations of the candidate vaccine and were then monitored for safety and reactogenicity for four weeks after each dose, and for their immune responses up to 28 weeks. RESULTS Of 36 adults enrolled, 35 completed the study as planned. Either no or mild adverse events were observed, mainly injection site pain and tiredness. Seroconversion was 100% after two vaccinations. High geometric mean neutralizing antibody titers (GMT) were observed 14 days post first dose, peaking 14 days post second dose (at Day 42) in both high and low dose groups; GMTs on days 14, 28, 42, and 56 were 128, 81, 323, 203 and 144, 100, 451, 351 in low- and high-dose groups, respectively. Titers for both doses declined gradually to Day 196 but remained higher than baseline and the placebo groups, which had low GMTs throughout the duration of the study. Cross-neutralizing antibody activity against heterologous sub-genotypes was demonstrated. CONCLUSION These data show that the EV71 candidate vaccine is safe and immunogenic in adults and supports further clinical development as a potential pediatric vaccine by initiating a dose-escalation study for determining the dose-dependent safety and immunogenicity of the vaccine in young naïve children.
Collapse
Affiliation(s)
- Paul A Tambyah
- Department of Medicine, NUH Investigational Medicine Unit, Yong Loo Lin School of Medicine, National University of Singapore, 1E, Kent Ridge Road, NUHS Tower Block, Level 10, Singapore 119228, Singapore
| | - Jolene Oon
- Department of Medicine, NUH Investigational Medicine Unit, Yong Loo Lin School of Medicine, National University of Singapore, 1E, Kent Ridge Road, NUHS Tower Block, Level 10, Singapore 119228, Singapore
| | - Rosmonaliza Asli
- Department of Medicine, NUH Investigational Medicine Unit, Yong Loo Lin School of Medicine, National University of Singapore, 1E, Kent Ridge Road, NUHS Tower Block, Level 10, Singapore 119228, Singapore
| | - William Kristanto
- Department of Medicine, NUH Investigational Medicine Unit, Yong Loo Lin School of Medicine, National University of Singapore, 1E, Kent Ridge Road, NUHS Tower Block, Level 10, Singapore 119228, Singapore
| | - Shi-Hsia Hwa
- Vaccine Business Unit, Takeda Pharmaceuticals Asia Pacific Pte Ltd, 21 Biopolis Road, Nucleos South Tower Level 4, Singapore 138567, Singapore
| | - Fue Vang
- Vaccine Business Unit, Takeda Pharmaceuticals USA, 40 Landsdowne Street, Cambridge, MA 02139, USA
| | - Lovkesh Karwal
- Vaccine Business Unit, Takeda Pharmaceuticals USA, 40 Landsdowne Street, Cambridge, MA 02139, USA
| | - Jeremy Fuchs
- Vaccine Business Unit, Takeda Pharmaceuticals USA, 40 Landsdowne Street, Cambridge, MA 02139, USA
| | - Joseph D Santangelo
- Vaccine Business Unit, Takeda Pharmaceuticals USA, 40 Landsdowne Street, Cambridge, MA 02139, USA
| | - Gilad S Gordon
- Takeda Vaccines, Inc., Takeda Pharmaceuticals USA, Fort Collins, CO, USA
| | - Cynthia Thomson
- Vaccine Business Unit, Takeda Pharmaceuticals Asia Pacific Pte Ltd, 21 Biopolis Road, Nucleos South Tower Level 4, Singapore 138567, Singapore
| | - Raman Rao
- Vaccine Business Unit, Takeda Pharmaceuticals Asia Pacific Pte Ltd, 21 Biopolis Road, Nucleos South Tower Level 4, Singapore 138567, Singapore
| | - Hansi Dean
- Vaccine Business Unit, Takeda Pharmaceuticals USA, 40 Landsdowne Street, Cambridge, MA 02139, USA
| | - Subash C Das
- Vaccine Business Unit, Takeda Pharmaceuticals USA, 40 Landsdowne Street, Cambridge, MA 02139, USA.
| | - Dan T Stinchcomb
- Takeda Vaccines, Inc., Takeda Pharmaceuticals USA, Fort Collins, CO, USA
| |
Collapse
|
14
|
Wang WH, Erazo EM, Ishcol MRC, Lin CY, Assavalapsakul W, Thitithanyanont A, Wang SF. Virus-induced pathogenesis, vaccine development, and diagnosis of novel H7N9 avian influenza A virus in humans: a systemic literature review. J Int Med Res 2019; 48:300060519845488. [PMID: 31068040 PMCID: PMC7140199 DOI: 10.1177/0300060519845488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
H7N9 avian influenza virus (AIV) caused human infections in 2013 in China.
Phylogenetic analyses indicate that H7N9 AIV is a novel reassortant strain with
pandemic potential. We conducted a systemic review regarding virus-induced
pathogenesis, vaccine development, and diagnosis of H7N9 AIV infection in
humans. We followed PRISMA guidelines and searched PubMed, Web of Science, and
Google Scholar to identify relevant articles published between January 2013 and
December 2018. Pathogenesis data indicated that H7N9 AIV belongs to low
pathogenic avian influenza, which is mostly asymptomatic in avian species;
however, H7N9 induces high mortality in humans. Sporadic human infections have
recently been reported, caused by highly pathogenic avian influenza viruses
detected in poultry. H7N9 AIVs resistant to adamantine and oseltamivir cause
severe human infection by rapidly inducing progressive acute community-acquired
pneumonia, multiorgan dysfunction, and cytokine dysregulation; however,
mechanisms via which the virus induces severe syndromes remain unclear. An H7N9
AIV vaccine is lacking; designs under evaluation include synthesized peptide,
baculovirus-insect system, and virus-like particle vaccines. Molecular diagnosis
of H7N9 AIVs is suggested over conventional assays, for biosafety reasons.
Several advanced or modified diagnostic assays are under investigation and
development. We summarized virus-induced pathogenesis, vaccine development, and
current diagnostic assays in H7N9 AIVs.
Collapse
Affiliation(s)
- Wen-Hung Wang
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung.,Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung
| | - Esmeralda Merari Erazo
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung
| | - Max R Chang Ishcol
- Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung
| | - Chih-Yen Lin
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung
| | - Wanchai Assavalapsakul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | | | - Sheng-Fan Wang
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung.,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung
| |
Collapse
|
15
|
Haveri A, Ikonen N, Kantele A, Anttila VJ, Ruotsalainen E, Savolainen-Kopra C, Julkunen I. Seasonal influenza vaccines induced high levels of neutralizing cross-reactive antibody responses against different genetic group influenza A(H1N1)pdm09 viruses. Vaccine 2019; 37:2731-2740. [PMID: 30954308 DOI: 10.1016/j.vaccine.2019.03.078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/25/2019] [Accepted: 03/31/2019] [Indexed: 12/27/2022]
Abstract
Influenza A(H1N1)pdm09 viruses have been circulating throughout the world since the 2009 pandemic. A/California/07/2009 (H1N1) virus was included in seasonal influenza vaccines for seven years altogether, providing a great opportunity to analyse vaccine-induced immunity in relation to the postpandemic evolution of the A(H1N1)pdm09 virus. Serum antibodies against various epidemic strains of influenza A(H1N1)pdm09 viruses were measured among health care workers (HCWs) by haemagglutination inhibition and microneutralization tests before and after 2010 and 2012 seasonal influenza vaccinations. We detected high responses of vaccine-induced neutralizing antibodies to six distinct genetic groups. Our results indicate antigenic similarity between vaccine and circulating A(H1N1)pdm09 strains, and substantial vaccine-induced immunity against circulating epidemic viruses.
Collapse
MESH Headings
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immunity, Humoral
- Influenza A Virus, H1N1 Subtype/classification
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Neutralization Tests
- Phylogeny
- Seasons
- Serologic Tests
- Structure-Activity Relationship
- Vaccination
Collapse
Affiliation(s)
- Anu Haveri
- Expert Microbiology Unit, Department of Health Security, National Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland.
| | - Niina Ikonen
- Expert Microbiology Unit, Department of Health Security, National Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland
| | - Anu Kantele
- Division of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, POB 348, 00029 HUS Helsinki, Finland
| | - Veli-Jukka Anttila
- Division of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, POB 348, 00029 HUS Helsinki, Finland
| | - Eeva Ruotsalainen
- Division of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, POB 348, 00029 HUS Helsinki, Finland
| | - Carita Savolainen-Kopra
- Expert Microbiology Unit, Department of Health Security, National Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland
| | - Ilkka Julkunen
- Expert Microbiology Unit, Department of Health Security, National Institute for Health and Welfare (THL), POB 30, 00271 Helsinki, Finland; Institute of Biomedicine, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520 Turku, Finland
| |
Collapse
|
16
|
Gianchecchi E, Torelli A, Montomoli E. The use of cell-mediated immunity for the evaluation of influenza vaccines: an upcoming necessity. Hum Vaccin Immunother 2019; 15:1021-1030. [PMID: 30614754 PMCID: PMC6605831 DOI: 10.1080/21645515.2019.1565269] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Influenza vaccines are a fundamental tool for preventing the disease and reducing its consequences, particularly in specific high-risk groups. In order to be licensed, influenza vaccines have to meet strict criteria established by European Medicines Agency. Although the licensure of influenza vaccines started 65 years ago, Hemagglutination Inhibition and Single Radial Hemolysis are the only serological assays that can ascertain correlates of protection. However, they present evident limitations. The present review focuses on the evaluation of cell-mediated immunity (CMI), which plays an important role in the host immune response in protecting against virus-related illness and in the establishment of long-term immunological memory. Although correlates of protection are not currently available for CMI, it would be advisable to investigate this kind of immunological response for the evaluation of next-generation vaccines.
Collapse
Affiliation(s)
| | - A Torelli
- a VisMederi srl , Siena , Italy.,b Department of Life Sciences , University of Siena , Siena , Italy
| | - E Montomoli
- a VisMederi srl , Siena , Italy.,c Department of Molecular and Developmental Medicine , University of Siena , Siena , Italy
| |
Collapse
|
17
|
Shim DH, Kim MJ, Cha HR, Park ES, Kim AR, Park JH, Park HC, Song D, Lee JM. Development of a HA1-specific enzyme-linked immunosorbent assay against pandemic influenza virus A H1N1. Clin Exp Vaccine Res 2019; 8:70-76. [PMID: 30775353 PMCID: PMC6369127 DOI: 10.7774/cevr.2019.8.1.70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/21/2019] [Accepted: 01/25/2019] [Indexed: 12/02/2022] Open
Abstract
Purpose Enzyme-linked immunosorbent assay (ELISA) has been used in the diverse field to evaluate influenza virus infection; for the surveillance, diagnosis, efficacy evaluation, and development of the vaccine. The aim of this study was to establish an ELISA for detecting HA strain-specific antibodies using recombinant pandemic A H1N1 (pH1N1) HA1 (rHA1) protein. Materials and Methods rHA1 was produced in baculovirus system. The clinical performance of the developed ELISA was validated using human serum samples, by comparison with standard methods for detecting a neutralizing antibody; hemagglutination inhibition (HI) assay and microneutralization test (MNT). The ability of the ELISA system to evaluate the efficacy test of an influenza vaccine was explored by measuring antibody levels in the serum of vaccinated mice. Results Our ELISA could detect anti-rHA1 antibody in influenza-infected patients and vaccinated subjects. Compared to HI assay and MNT as reference methods, our method showed good performance in detection of anti-rHA1 antibody. Detection of the anti-rHA1 antibody in vaccinated mice and its correlation with titers in HI assay was also proved in a mice model. Conclusion An ELISA system using rHA1 of pH1N1 influenza virus was developed, and showed good clinical performance in diagnosis of influenza virus infection and evaluation of the vaccination efficacy in both human and animal models.
Collapse
Affiliation(s)
- Doo Hee Shim
- Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Min Jung Kim
- Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Pediatrics, Severance Hospital, Institute of Allergy, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hye-Ran Cha
- Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Sun Park
- Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ah Reum Kim
- Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jeon Han Park
- Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung-Cheon Park
- Department of Internal Medicine, Gangnam Severance Hospital, Institute of Vascular and Metabolism Research, Yonsei University College of Medicine, Seoul, Korea
| | - Daesub Song
- College of Pharmacy, Korea University, Sejong, Korea
| | - Jae Myun Lee
- Department of Microbiology and Immunology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Lee MS, Jang EY, Cho J, Kim K, Lee CH, Yi H. Development and comparison of two H5N8 influenza A vaccine candidate strains. Arch Virol 2018; 164:127-136. [PMID: 30291503 DOI: 10.1007/s00705-018-4062-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 09/15/2018] [Indexed: 11/28/2022]
Abstract
Avian influenza viruses circulating in birds have caused outbreaks of infection in poultry and humans, thereby threatening public health. Recently, a highly pathogenic avian influenza (HPAI) virus (H5N8) of clade 2.3.4.4 emerged in Korea and other countries and caused multiple outbreaks in domestic and wild birds, with concerns for human infection. To combat HPAI viral infections, novel vaccines are likely to be the most effective approach. Therefore, in this study, we generated H5N8 vaccine candidate viruses based on a Korean isolate (A/broiler duck/Korea/Buan2/2014). The vaccine candidate viruses were 2:6 reassortants expressing the two surface glycoproteins of A/broiler duck/Korea/Buan2/2014 on an A/Puerto Rico/8/34 (PR8) backbone generated by using an eight-plasmid-based reverse genetics system with or without replacement of the multi-basic amino acid cleavage motif (MBCM, a crucial pathogenic factor in HPAI virus) with a bi-basic amino acid cleavage motif (BBCM) in their HA. An H5N8 vaccine candidate virus containing the BBCM showed attenuated pathogenesis in embryonated eggs and exhibited less virulence in the infected mice compared with the wild H5N8 virus containing an MBCM. Vaccination with an inactivated preparation of the vaccine candidate virus protected mice from lethal H5N8 viral challenge. This is the first report of the development and evaluation of H5N8 vaccine strains (with an MBCM or BBCM) of HA clade 2.3.4.4 as vaccine candidates. Our findings suggest that H5N8 strains with a BBCM instead of an MBCM might be considered for H5N8 vaccine seed virus development or as a reference vaccine against H5N8 viral strains.
Collapse
Affiliation(s)
- Mi-Seon Lee
- Division of Viral Disease Research, Center for Infectious Diseases Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, 187, Osongsaengmyeong2-ro, Cheongju-si, 28159, Chungcheongbuk-do, Korea.,Department of Life Science and Technology, Pai Chai University, Daejeon, Korea
| | - Eun Young Jang
- Division of Viral Disease Research, Center for Infectious Diseases Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, 187, Osongsaengmyeong2-ro, Cheongju-si, 28159, Chungcheongbuk-do, Korea.,Department of Microbiology, Chungbuk National University, Cheongju, Korea
| | - Junhyung Cho
- Division of Viral Disease Research, Center for Infectious Diseases Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, 187, Osongsaengmyeong2-ro, Cheongju-si, 28159, Chungcheongbuk-do, Korea.,Department of Microbiology, Chungbuk National University, Cheongju, Korea
| | - Kisoon Kim
- Division of Viral Disease Research, Center for Infectious Diseases Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, 187, Osongsaengmyeong2-ro, Cheongju-si, 28159, Chungcheongbuk-do, Korea
| | - Chan Hee Lee
- Department of Microbiology, Chungbuk National University, Cheongju, Korea
| | - Hwajung Yi
- Division of Viral Disease Research, Center for Infectious Diseases Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, 187, Osongsaengmyeong2-ro, Cheongju-si, 28159, Chungcheongbuk-do, Korea.
| |
Collapse
|
19
|
Moreno A, Lelli D, Lavazza A, Sozzi E, Zanni I, Chiapponi C, Foni E, Capucci L, Brocchi E. MAb‐based competitive ELISA for the detection of antibodies against influenza D virus. Transbound Emerg Dis 2018; 66:268-276. [DOI: 10.1111/tbed.13012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/11/2018] [Accepted: 08/28/2018] [Indexed: 02/02/2023]
Affiliation(s)
- Ana Moreno
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Brescia Italy
| | - Davide Lelli
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Brescia Italy
| | - Antonio Lavazza
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Brescia Italy
| | - Enrica Sozzi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Brescia Italy
| | - Irene Zanni
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Parma Italy
| | - Chiara Chiapponi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Parma Italy
| | - Emanuela Foni
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Parma Italy
| | - Lorenzo Capucci
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Brescia Italy
| | - Emiliana Brocchi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna Brescia Italy
| |
Collapse
|
20
|
Zhao N, Grund C, Beer M, Harder TC. Engineered recombinant protein products of the avian paramyxovirus type-1 nucleocapsid and phosphoprotein genes for serological diagnosis. Virol J 2018; 15:8. [PMID: 29325564 PMCID: PMC5765633 DOI: 10.1186/s12985-018-0924-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/08/2018] [Indexed: 11/29/2022] Open
Abstract
Background Virulent Newcastle disease virus (NDV, avian Avulavirus-1, APMV-1) induces a highly contagious and lethal systemic disease in gallinaceous poultry. APMV-1 antibody detection is used for surveillance and to control vaccination, but is hampered by cross-reactivity to other subtypes of avian Avulaviruses. Data are lacking concerning the applicability of NDV V proteins as differential diagnostic marker to distinguish vaccinated from virus-infected birds (DIVA strategy). Methods Full length and C-terminally truncated nucleocapsid (NP) protein, and the unique C-terminal regions of the phospho- (P) and V proteins of the NDV LaSota strain were bacterially expressed as fusion proteins with the multimerization domain of the human C4 binding protein, and used as diagnostic antigens in indirect ELISA. Results When used as diagnostic antigen in indirect ELISAs, recombinant full-length proved to be a sensitive target to detect seroconversion in chickens after APMV-1 vaccination and infection, but revealed some degree of cross reactivity with sera raised against other APMV subtypes. Cross reactivity was abolished but also sensitivity decreased when employing a C-terminal fragment of the NP of NDV as diagnostic antigen. Antibodies to the NDV V protein were mounted in poultry following NDV infection but also, albeit at lower rates and titers, after vaccination with attenuated NDV vaccines. V-specific seroconversion within the flock was incomplete and titers in individual bird transient. Conclusions Indirect ELISA based on bacterially expressed recombinant full-length NP compared favorably with a commercial NDV ELISA based on whole virus antigen, but cross reactivity between the NP proteins of different APMV subtypes could compromise specificity. However, specificity increased when using a less conserved C-terminal fragment of NP instead. Moreover, a serological DIVA strategy built on the NDV V protein was not feasible due to reduced immunogenicity of the V protein and frequent use of live-attenuated NDV vaccines. Electronic supplementary material The online version of this article (10.1186/s12985-018-0924-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Na Zhao
- The Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut, Institute of Diagnostic Virology, Suedufer 10, 17493, Greifswald, Germany
| | - Christian Grund
- The Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut, Institute of Diagnostic Virology, Suedufer 10, 17493, Greifswald, Germany
| | - Martin Beer
- The Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut, Institute of Diagnostic Virology, Suedufer 10, 17493, Greifswald, Germany
| | - Timm C Harder
- The Federal Research Institute for Animal Health, Friedrich-Loeffler-Institut, Institute of Diagnostic Virology, Suedufer 10, 17493, Greifswald, Germany.
| |
Collapse
|
21
|
Yuan HY, Baguelin M, Kwok KO, Arinaminpathy N, van Leeuwen E, Riley S. The impact of stratified immunity on the transmission dynamics of influenza. Epidemics 2017; 20:84-93. [PMID: 28395850 PMCID: PMC5628170 DOI: 10.1016/j.epidem.2017.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 03/02/2017] [Accepted: 03/08/2017] [Indexed: 12/09/2022] Open
Abstract
The disease model with stratified immunity improves the accuracy on influenza epidemic reconstruction. Antibody boosting in children is greater than adults during influenza outbreak. Age-specific mixing pattern and the relative infectivity of children to adults are the key drivers of infection heterogeneity.
Although empirical studies show that protection against influenza infection in humans is closely related to antibody titres, influenza epidemics are often described under the assumption that individuals are either susceptible or not. Here we develop a model in which antibody titre classes are enumerated explicitly and mapped onto a variable scale of susceptibility in different age groups. Fitting only with pre- and post-wave serological data during 2009 pandemic in Hong Kong, we demonstrate that with stratified immunity, the timing and the magnitude of the epidemic dynamics can be reconstructed more accurately than is possible with binary seropositivity data. We also show that increased infectiousness of children relative to adults and age-specific mixing are required to reproduce age-specific seroprevalence observed in Hong Kong, while pre-existing immunity in the elderly is not. Overall, our results suggest that stratified immunity in an aged-structured heterogeneous population plays a significant role in determining the shape of influenza epidemics.
Collapse
Affiliation(s)
- Hsiang-Yu Yuan
- MRC Centre for Outbreak Analysis and Disease Modelling, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| | - Marc Baguelin
- Respiratory Diseases Department, Public Health England, London, United Kingdom; Centre for the Mathematical Modelling of Infectious Disease, Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom.
| | - Kin O Kwok
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Nimalan Arinaminpathy
- MRC Centre for Outbreak Analysis and Disease Modelling, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| | - Edwin van Leeuwen
- MRC Centre for Outbreak Analysis and Disease Modelling, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom; Respiratory Diseases Department, Public Health England, London, United Kingdom
| | - Steven Riley
- MRC Centre for Outbreak Analysis and Disease Modelling, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom.
| |
Collapse
|
22
|
Preexisting Immunity, Not Frailty Phenotype, Predicts Influenza Postvaccination Titers among Older Veterans. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00498-16. [PMID: 28100496 DOI: 10.1128/cvi.00498-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/10/2017] [Indexed: 01/08/2023]
Abstract
Both preexisting immunity to influenza and age have been shown to be correlates of influenza vaccine responses. Frailty, an indicator of functional impairment in older adults, was also shown in one study to predict lower influenza vaccine responses among nonveterans. In the current study, we aimed to determine the associations between frailty, preexisting immunity, and immune responses to influenza vaccine among older veterans. We studied 117 subjects (age range, 62 to 95 years [median age, 81 years]), divided into three cohorts based on the Fried frailty test, i.e., nonfrail (NF) (n = 23 [median age, 68 years]), prefrail (n = 50 [median age, 80 years]), and frail (n = 44 [median age, 82 years]), during the 2010-2011 and 2011-2012 influenza seasons. Subjects received the seasonal trivalent inactivated influenza vaccine, and baseline and postvaccination samples were obtained. Anti-influenza humoral immunity, as measured by hemagglutination inhibition (HI) and microneutralization assays, was measured for influenza B, A(H1N1)pdm09, and A(H3N2) viruses. Postvaccination titers were not different between frail and NF subjects overall in this older subset of veterans. However, preexisting HI titers were strongly correlated with postvaccination titers among all functional status groups. When microneutralization titers were compared, the association between preexisting immunity and vaccine responses varied by frailty status, with the strongest correlation being observed for the NF group. In conclusion, preexisting immunity rather than frailty appeared to predict postvaccination titers in this older veteran cohort.
Collapse
|
23
|
Damle RG, Patil AA, Bhide VS, Pawar SD, Sapkal GN, Bondre VP. Development of a novel rapid micro-neutralization ELISA for the detection of neutralizing antibodies against Chandipura virus. J Virol Methods 2016; 240:1-6. [PMID: 27856212 DOI: 10.1016/j.jviromet.2016.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 11/11/2016] [Accepted: 11/11/2016] [Indexed: 12/01/2022]
Abstract
BACKGROUND Chandipura virus (CHPV) is a leading cause of acute encephalitis with high mortality in paediatric population in India. A micro-neutralization ELISA (MN-ELISA) assay was developed for the detection of neutralizing antibodies (Nab) against CHPV. This novel method gives read-out in the form of ELISA optical density (OD) values and has a shorter turn-around time (TAT) as compared to the conventional cytopathic effect (CPE)-based neutralization assay (MN-CPE). The assay was developed using an Indian strain of CHPV. During the development of the assay different parameters such as cell count, dilution of primary and secondary antibodies and time point for the test termination were optimized. The new and conventional assays were run in parallel where known positive and negative human serum samples were used as test controls. The conventional MN-CPE was terminated at 48h post-infection (p.i.) and stained with Amido black, while in the new assay, MN-ELISA was terminated at pre-determined 18h p.i. and the infected cells were fixed with acetone, followed by in-situ ELISA. Results of both the assays were compared. The sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) of the new test was 100% when compared with the conventional MN-CPE method as a 'gold standard'. The MN-ELISA showed two-fold higher antibody titer in one sample and one sample was additionally positive than MN-CPE ELISA. CONCLUSION The MN-ELISA is rapid, more sensitive and read-out of results is by measurement of OD, which could be more accurate than manual observation of reduction in CPE. This novel test could be used as an alternative to the conventional MN-CPE based assay in sero-surveillance and in future vaccine studies.
Collapse
Affiliation(s)
- R G Damle
- National Institute of Virology, Pune, Maharashtra, India.
| | - A A Patil
- National Institute of Virology, Pune, Maharashtra, India
| | - V S Bhide
- National Institute of Virology, Pune, Maharashtra, India
| | - S D Pawar
- National Institute of Virology, Pune, Maharashtra, India
| | - G N Sapkal
- National Institute of Virology, Pune, Maharashtra, India
| | - V P Bondre
- National Institute of Virology, Pune, Maharashtra, India
| |
Collapse
|
24
|
Seasonal influenza vaccines and hurdles to mutual protection. Clin Microbiol Infect 2016; 22 Suppl 5:S113-S119. [PMID: 27568914 DOI: 10.1016/j.cmi.2016.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/19/2016] [Indexed: 11/22/2022]
Abstract
While vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent epidemiologic data question the appropriateness of traditional vaccination timing (prior to the winter season) in many parts of the world. Furthermore, vaccine uptake in most countries even in high-risk populations does not reach the 75% target recommended by the World Health Organization. Influenza viruses continually undergo antigenic variation, and both inactivated and live attenuated influenza vaccines confer only short-lived strain-specific immunity, so annual revaccination is required. Improving vaccine-induced immunity is therefore an important goal. A vaccine that could confer durable protection against emerging influenza strains could significantly reduce onward transmission. Therefore, further understanding of protective immunity against influenza (including broadly cross-protective immune mechanisms such as haemagglutinin stem-binding antibodies and T cells) offers the hope of vaccines that can confer the long-lived heterosubtypic immune responses required for mutual protection.
Collapse
|
25
|
Gupta P, Sharma A, Spurgers KB, Bakken RR, Eccleston LT, Cohen JW, Honnold SP, Glass PJ, Maheshwari RK. 1,5-Iodonaphthyl azide-inactivated V3526 protects against aerosol challenge with virulent venezuelan equine encephalitis virus. Vaccine 2016; 34:2762-5. [PMID: 27129427 DOI: 10.1016/j.vaccine.2016.04.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/06/2016] [Accepted: 04/10/2016] [Indexed: 10/21/2022]
Abstract
Venezuelan equine encephalitis virus (VEEV) is a New World alphavirus. VEEV is highly infectious in aerosolized form and has been identified as a bio-terrorism agent. There is no licensed vaccine for prophylaxis against VEEV. The current IND vaccine is poorly immunogenic and does not protect against an aerosol challenge with virulent VEEV. We have previously shown that VEEV inactivated by 1,5-iodonaphthyl azide (INA) protects against footpad challenge with virulent VEEV. In this study, we inactivated an attenuated strain of VEEV, V3526, with INA and evaluated its protective efficacy against aerosol challenge with wild type VEEV. We demonstrated that among three routes of immunization, intramuscular immunization with INA-inactivate V3526 (INA-iV3526) provided complete protection against aerosol challenge with virulent VEEV. Our data suggests that INA-iV3526 can be explored further for development as an effective vaccine candidate against aerosol challenge of virulent VEEV.
Collapse
Affiliation(s)
- Paridhi Gupta
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | - Anuj Sharma
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States.
| | - Kevin B Spurgers
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Russell R Bakken
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Lori T Eccleston
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Jeffrey W Cohen
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Shelley P Honnold
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Pamela J Glass
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Radha K Maheshwari
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| |
Collapse
|
26
|
Grund S, Michel S, Barthuber C, Adams O. Serum and mucosal antibodies fail as prognostic markers during critical influenza A infection. J Clin Virol 2015; 74:32-6. [PMID: 26655265 DOI: 10.1016/j.jcv.2015.11.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/12/2015] [Accepted: 11/15/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND Previous studies have indicated that the absence of serum antibodies to influenza A H1N1 virus on day 4 after onset of symptoms predicted a fatal outcome in patients critically ill with influenza. The underlying mechanism was suggested to be the trapping of anti-influenza antibodies in pulmonary immune complexes. OBJECTIVES To study serum and mucosal antibodies as prognostic markers in patients with severe influenza A H1N1 infection. STUDY DESIGN Blood and respiratory samples (n=324) from 12 patients with severe influenza were analysed for anti-H1N1 antibodies with and without immune complex dissociation from symptom onset until convalescence or death (follow up 14-169 days). Eleven healthy subjects were analysed for comparison. RESULTS One of the 12 patients died from influenza pneumonia and had no detectable anti-H1N1 serum antibodies. However, also 2 of the 11 surviving patients remained negative for anti-H1N1 serum antibodies during follow-up (20 and 41 days, respectively). In six of the 11 survivors serum antibodies on day 4 were negative, but turned positive between day 7 and 23. In the remaining 3 patients antibodies were detected in the first 4 days of illness. Mucosal IgG or IgA was detected in all of the patients regardless of their clinical outcome and in 4 of 11 healthy subjects. No mucosal immune complexes were found in the patient who died but were detected in 3 of the 11 survivors. CONCLUSIONS This study suggests that no prognostic conclusions can be drawn from anti-H1N1 serum and mucosal antibodies in patients with severe influenza.
Collapse
Affiliation(s)
- Sebastian Grund
- University of Düsseldorf, Medical Faculty, Institute for Virology, D-40225 Düsseldorf, Germany.
| | - Stefan Michel
- University of Düsseldorf, Medical Faculty, Institute for Virology, D-40225 Düsseldorf, Germany
| | - Carmen Barthuber
- University of Düsseldorf, Medical Faculty, Institute for Clinical Chemistry, D-40225 Düsseldorf, Germany
| | - Ortwin Adams
- University of Düsseldorf, Medical Faculty, Institute for Virology, D-40225 Düsseldorf, Germany
| |
Collapse
|
27
|
Kay AW, Bayless NL, Fukuyama J, Aziz N, Dekker CL, Mackey S, Swan GE, Davis MM, Blish CA. Pregnancy Does Not Attenuate the Antibody or Plasmablast Response to Inactivated Influenza Vaccine. J Infect Dis 2015; 212:861-70. [PMID: 25740957 PMCID: PMC4548461 DOI: 10.1093/infdis/jiv138] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/26/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Inactivated influenza vaccine (IIV) is recommended during pregnancy to prevent influenza infection and its complications in pregnant women and their infants. However, the extent to which pregnancy modifies the antibody response to vaccination remains unclear, and prior studies have focused primarily on hemagglutinin inhibition (HI) titers. A more comprehensive understanding of how pregnancy modifies the humoral immune response to influenza vaccination will aid in maximizing vaccine efficacy. METHODS Healthy pregnant women and control women were studied prior to, 7 days after, and 28 days after vaccination with IIV. HI titers, microneutralization (MN) titers, and the frequency of circulating plasmablasts were evaluated in pregnant versus control women. RESULTS Pregnant women and control women mount similarly robust serologic immune responses to IIV, with no significant differences for any influenza strain in postvaccination geometric mean HI or MN titers. HI and MN titers correlate, though MN titers demonstrate more robust changes pre- versus postvaccination. The induction of circulating plasmablasts is increased in pregnant women versus controls (median fold-change 2.60 vs 1.49 [interquartile range, 0.94-7.53 vs 0.63-2.67]; P = .03). CONCLUSIONS Pregnant women do not have impaired humoral immune responses to IIV and may have increased circulating plasmablast production compared to control women.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gary E. Swan
- Stanford Prevention Research Center, Department of Medicine
| | - Mark M. Davis
- Stanford Immunology, Stanford University School of Medicine
- Department of Microbiology & Immunology
| | - Catherine A. Blish
- Stanford Immunology, Stanford University School of Medicine
- Department of Medicine, Stanford University School of Medicine, California
| |
Collapse
|
28
|
Verschoor CP, Singh P, Russell ML, Bowdish DME, Brewer A, Cyr L, Ward BJ, Loeb M. Microneutralization assay titres correlate with protection against seasonal influenza H1N1 and H3N2 in children. PLoS One 2015; 10:e0131531. [PMID: 26107625 PMCID: PMC4479562 DOI: 10.1371/journal.pone.0131531] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/03/2015] [Indexed: 12/27/2022] Open
Abstract
Although the microneutralization (MN) assay has been shown to be more sensitive than the hemagglutination inhibition (HAI) assay for the measurement of humoral immunity against influenza viruses, further evidence relating MN titres to protective efficacy against infection is needed. Serum antibodies against seasonal H1N1 and H3N2 influenza were measured in children and adolescents (n = 656) by MN and hemagglutination inhibition (HAI) assays. Compared to HAI, the MN assay is more sensitive in detecting serum antibodies and estimates of protective effectiveness against PCR-confirmed infection were higher for both subtypes. Given our findings, the MN assay warrants further consideration as a formal tool for the routine evaluation of vaccine-induced antibody responses.
Collapse
Affiliation(s)
- Chris P. Verschoor
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Michael G DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Pardeep Singh
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | | | - Dawn M. E. Bowdish
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Michael G DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Angela Brewer
- Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Louis Cyr
- Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Brian J. Ward
- Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Mark Loeb
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Michael G DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
29
|
He W, Mullarkey CE, Miller MS. Measuring the neutralization potency of influenza A virus hemagglutinin stalk/stem-binding antibodies in polyclonal preparations by microneutralization assay. Methods 2015; 90:95-100. [PMID: 25957674 DOI: 10.1016/j.ymeth.2015.04.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 11/17/2022] Open
Abstract
The discovery of broadly-neutralizing antibodies that bind to the hemagglutinin stalk/stem domain has opened exciting new avenues for the development of "universal" influenza virus vaccines and therapeutics. Unlike strain-specific antibodies which bind to the hemagglutinin head domain and inhibit receptor binding, antibodies that bind to the stalk domain function to inhibit later stages of infection. The hemagglutination inhibition assay has long been the standard for evaluating titers of neutralizing hemagglutinin-specific antibodies in serum. The assay has the beneficial properties of being relatively rapid, easy-to-perform, and requires very little specialized equipment. Historically, hemagglutination inhibition titers of 40 or above against a given strain of influenza has been considered a correlate of protection on a population level. Unfortunately, this assay cannot be used to measure titers of hemagglutinin stalk-specific antibodies due to their lack of hemagglutination inhibiting activity. This has necessitated the development of novel reagents and assays capable of sensitive and specific detection of broadly-neutralizing HA stalk-binding antibodies in polyclonal mixtures. Here, we describe a novel microneutralization-based assay that utilizes recombinant influenza A viruses expressing chimeric hemagglutinin molecules and 'exotic' neuraminidase to measure titers of broadly-neutralizing antibodies in polyclonal preparations.
Collapse
Affiliation(s)
- Wenqian He
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Caitlin E Mullarkey
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew S Miller
- Department of Biochemistry and Biomedical Sciences, Institute for Infectious Diseases Research, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
30
|
Kucharski AJ, Lessler J, Read JM, Zhu H, Jiang CQ, Guan Y, Cummings DAT, Riley S. Estimating the life course of influenza A(H3N2) antibody responses from cross-sectional data. PLoS Biol 2015; 13:e1002082. [PMID: 25734701 PMCID: PMC4348415 DOI: 10.1371/journal.pbio.1002082] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 01/16/2015] [Indexed: 02/05/2023] Open
Abstract
The immunity of a host population against specific influenza A strains can influence a number of important biological processes, from the emergence of new virus strains to the effectiveness of vaccination programmes. However, the development of an individual's long-lived antibody response to influenza A over the course of a lifetime remains poorly understood. Accurately describing this immunological process requires a fundamental understanding of how the mechanisms of boosting and cross-reactivity respond to repeated infections. Establishing the contribution of such mechanisms to antibody titres remains challenging because the aggregate effect of immune responses over a lifetime are rarely observed directly. To uncover the aggregate effect of multiple influenza infections, we developed a mechanistic model capturing both past infections and subsequent antibody responses. We estimated parameters of the model using cross-sectional antibody titres to nine different strains spanning 40 years of circulation of influenza A(H3N2) in southern China. We found that "antigenic seniority" and quickly decaying cross-reactivity were important components of the immune response, suggesting that the order in which individuals were infected with influenza strains shaped observed neutralisation titres to a particular virus. We also obtained estimates of the frequency and age distribution of influenza infection, which indicate that although infections became less frequent as individuals progressed through childhood and young adulthood, they occurred at similar rates for individuals above age 30 y. By establishing what are likely to be important mechanisms driving epochal trends in population immunity, we also identified key directions for future studies. In particular, our results highlight the need for longitudinal samples that are tested against multiple historical strains. This could lead to a better understanding of how, over the course of a lifetime, fast, transient antibody dynamics combine with the longer-term immune responses considered here.
Collapse
Affiliation(s)
- Adam J. Kucharski
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- * E-mail:
| | - Justin Lessler
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jonathan M. Read
- Department of Epidemiology and Population Health, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Huachen Zhu
- International Institute of Infection and Immunity, Shantou University Medical College, Shantou, Guangdong, China
- State Key Laboratory of Emerging Infectious Diseases and Centre of Influenza Research, University of Hong Kong, Hong Kong SAR, China
| | | | - Yi Guan
- International Institute of Infection and Immunity, Shantou University Medical College, Shantou, Guangdong, China
- State Key Laboratory of Emerging Infectious Diseases and Centre of Influenza Research, University of Hong Kong, Hong Kong SAR, China
| | - Derek A. T. Cummings
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Steven Riley
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
31
|
Takeda S, Hisano M, Komano J, Yamamoto H, Sago H, Yamaguchi K. Influenza vaccination during pregnancy and its usefulness to mothers and their young infants. J Infect Chemother 2015; 21:238-46. [PMID: 25708925 DOI: 10.1016/j.jiac.2015.01.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 01/20/2015] [Accepted: 01/28/2015] [Indexed: 11/29/2022]
Abstract
The current approach to protecting pregnant women from influenza infection and serious influenza-related complications is vaccination. It is, therefore, critical to evaluate the vaccine's safety, immunogenicity, and protection efficacy during pregnancy. However, because it is affected by previous influenza vaccination or infection, the efficacy of the seasonal trivalent inactivated influenza vaccine is difficult to evaluate in pregnant women. The A/H1N1pdm pandemic in 2009 provided us with the opportunity to evaluate the immunogenicity of the influenza vaccine unaffected by previous vaccinations or infections. Vaccination with inactivated influenza virus during pregnancy elicited neutralizing antibody titers that were sufficient and comparable to those of naturally infected individuals. Furthermore, post-pandemic surveys provided a wealth of definitive information on vaccine efficacy and safety. In addition, transplacental transfer of antibodies following vaccination protected newborn infants against influenza infection. With reports showing the effectiveness of influenza vaccine during pregnancy, it is suggested that influenza vaccination benefits both mothers and their young infants.
Collapse
Affiliation(s)
- Satoshi Takeda
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Michi Hisano
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Jun Komano
- Department of Infectious Diseases, Osaka Prefectural Institute of Public Health, Virology Division, 3-69, Nakamichi 1-chome, Higashinari-ku, Osaka 537-0025, Japan
| | - Hiroyuki Yamamoto
- AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Haruhiko Sago
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Koushi Yamaguchi
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| |
Collapse
|
32
|
Martin BE, Jia K, Sun H, Ye J, Hall C, Ware D, Wan XF. Detection of influenza antigenic variants directly from clinical samples using polyclonal antibody based proximity ligation assays. Virology 2015; 476:151-158. [PMID: 25546251 PMCID: PMC4902327 DOI: 10.1016/j.virol.2014.11.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/20/2014] [Accepted: 11/28/2014] [Indexed: 11/30/2022]
Abstract
Identification of antigenic variants is the key to a successful influenza vaccination program. The empirical serological methods to determine influenza antigenic properties require viral propagation. Here a novel quantitative PCR-based antigenic characterization method using polyclonal antibody and proximity ligation assays, or so-called polyPLA, was developed and validated. This method can detect a viral titer that is less than 1000 TCID50/mL. Not only can this method differentiate between different HA subtypes of influenza viruses but also effectively identify antigenic drift events within the same HA subtype of influenza viruses. Applications in H3N2 seasonal influenza data showed that the results from this novel method are consistent with those from the conventional serological assays. This method is not limited to the detection of antigenic variants in influenza but also other pathogens. It has the potential to be applied through a large-scale platform in disease surveillance requiring minimal biosafety and directly using clinical samples.
Collapse
MESH Headings
- Antibodies/analysis
- Antibodies, Viral/analysis
- Antigenic Variation
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- China
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- High-Throughput Screening Assays/instrumentation
- High-Throughput Screening Assays/methods
- Humans
- Influenza A Virus, H3N2 Subtype/classification
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/isolation & purification
- Influenza, Human/virology
- Polymerase Chain Reaction/instrumentation
- Polymerase Chain Reaction/methods
Collapse
Affiliation(s)
- Brigitte E Martin
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Kun Jia
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Hailiang Sun
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Jianqiang Ye
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Crystal Hall
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA
| | - Daphne Ware
- Mississippi Public Health Laboratory, Jackson, MS 39216, USA
| | - Xiu-Feng Wan
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA.
| |
Collapse
|
33
|
Yi H, Lee MS, Lee JY, Lee HK, Kang C. Immunological characterization of monoclonal antibodies used in rapid influenza diagnostic test for detection of the 2009 pandemic influenza A(H1N1)pdm09 infection. J Microbiol 2015; 53:166-75. [PMID: 25626373 DOI: 10.1007/s12275-015-4642-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/12/2015] [Accepted: 01/12/2015] [Indexed: 12/15/2022]
Abstract
Since the 2009 pandemic, monoclonal antibodies (mAbs) for rapid influenza diagnostic tests (RIDT) have been developed for specific diagnostics of pandemic viral infection. Most of the mAbs were poorly characterized because of urgency during the pandemic. Further characterization of the mAbs for RIDTs would be beneficial for understanding the immunological properties of the pandemic virus and utilizing the mAbs for other research purposes. In this study, it was confirmed that two mAbs (I38 and D383) in an RIDT for H1N1pdm09 diagnostics were able to detect H1N1pdm09 virus through enzyme-linked immunosorbent assay (ELISA) and immunofluorescence assay (IFA). Also, the two mAbs exhibited reactivity to hemagglutinins (HAs) of both the H1N1pdm09 and 1918 H1N1 viruses; therefore, the RIDT using the mAbs could detect HAs of H1N1pdm09 and also HAs of 1918 H1N1-like strains. In an extension to our previous study, the epitopes (Sa antigenic site and the interface area of F' and vestigial esterase subdomains on the HA1 domain of HA of H1N1pdm09) recognized by the mAbs were corroborated in depth by IFA with escape-mutants from the mAbs and mapping of the epitopes on the crystal structure of human H1N1 viral HAs. Collectively, these results imply that the mAbs for the RIDT may be suitable for use in studying the immunological properties of H1N1pdm09 viruses and that the Sa antigenic site and the interface area between F' and vestigial esterase subdomains on influenza viral HA recognized by the mAbs are immunologically conserved regions between H1N1pdm09 and 1918 H1N1.
Collapse
Affiliation(s)
- Hwajung Yi
- Division of Influenza Virus, Center for Infectious Diseases, Korea National Institute of Health, Centers for Disease Control and Prevention, Cheongju, 363-951, Republic of Korea,
| | | | | | | | | |
Collapse
|
34
|
Chiu C, Openshaw PJ. Antiviral B cell and T cell immunity in the lungs. Nat Immunol 2015; 16:18-26. [PMID: 25521681 PMCID: PMC7097128 DOI: 10.1038/ni.3056] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/14/2014] [Indexed: 12/13/2022]
Abstract
Respiratory viruses are frequent causes of repeated common colds, bronchitis and pneumonia, which often occur unpredictably as epidemics and pandemics. Despite those decimating effects on health and decades of intensive research, treatments remain largely supportive. The only commonly available vaccines are against influenza virus, and even these need improvement. The lung shares some features with other mucosal sites, but preservation of its especially delicate anatomical structures necessitates a fine balance of pro- and anti-inflammatory responses; well-timed, appropriately placed and tightly regulated T cell and B cell responses are essential for protection from infection and limitation of symptoms, whereas poorly regulated inflammation contributes to tissue damage and disease. Recent advances in understanding adaptive immunity should facilitate vaccine development and reduce the global effect of respiratory viruses.
Collapse
Affiliation(s)
- Christopher Chiu
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter J Openshaw
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
35
|
Sensing strategies for influenza surveillance. Biosens Bioelectron 2014; 61:357-69. [DOI: 10.1016/j.bios.2014.05.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 04/12/2014] [Accepted: 05/11/2014] [Indexed: 01/06/2023]
|
36
|
Abstract
Host immunity is a major driver of pathogen evolution and thus a major determinant of pathogen diversity. Explanations for pathogen diversity traditionally assume simple interactions between pathogens and the immune system, a view encapsulated by the susceptible-infected-recovered (SIR) model. However, there is growing evidence that the complexity of many host-pathogen interactions is dynamically important. This revised perspective requires broadening the definition of a pathogen's immunological phenotype, or what can be thought of as its immunological niche. After reviewing evidence that interactions between pathogens and host immunity drive much of pathogen evolution, I introduce the concept of a pathogen's immunological phenotype. Models that depart from the SIR paradigm demonstrate the utility of this perspective and show that it is particularly useful in understanding vaccine-induced evolution. This paper highlights questions in immunology, evolution, and ecology that must be answered to advance theories of pathogen diversity.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois
| |
Collapse
|
37
|
Li J, Arévalo MT, Chen Y, Chen S, Zeng M. T-cell-mediated cross-strain protective immunity elicited by prime-boost vaccination with a live attenuated influenza vaccine. Int J Infect Dis 2014; 27:37-43. [PMID: 25172265 DOI: 10.1016/j.ijid.2014.05.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/24/2014] [Accepted: 05/19/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Antigenic drift and shift of influenza viruses require frequent reformulation of influenza vaccines. In addition, seasonal influenza vaccines are often mismatched to the epidemic influenza strains. This stresses the need for a universal influenza vaccine. METHODS BALB/c mice were vaccinated with the trivalent live attenuated (LAIV; FluMist) or inactivated (TIV; FluZone) influenza vaccines and challenged with PR8 (H1N1), FM/47 (H1N1), or HK/68 (H3N2) influenza virus. Cytokines and antibody responses were tested by ELISA. Furthermore, different LAIV dosages were applied in BALB/c mice. LAIV vaccinated mice were also depleted of T-cells and challenged with PR8 virus. RESULTS LAIV induced significant protection against challenge with the non-vaccine strain PR8 influenza virus. Furthermore, protective immunity against PR8 was dose-dependent. Of note, interleukin 2 and interferon gamma cytokine secretion in the lung alveolar fluid were significantly elevated in mice vaccinated with LAIV. Moreover, T-cell depletion of LAIV vaccinated mice compromised protection, indicating that T-cell-mediated immunity is required. In contrast, passive transfer of sera from mice vaccinated with LAIV into naïve mice failed to protect against PR8 challenge. Neutralization assays in vitro confirmed that LAIV did not induce cross-strain neutralizing antibodies against PR8 virus. Finally, we showed that three doses of LAIV also provided protection against challenge with two additional heterologous viruses, FM/47 and HK/68. CONCLUSIONS These results support the potential use of the LAIV as a universal influenza vaccine under a prime-boost vaccination regimen.
Collapse
Affiliation(s)
- Junwei Li
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Maria T Arévalo
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Yanping Chen
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Shan Chen
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA
| | - Mingtao Zeng
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, 5001 El Paso Drive, El Paso, TX 79905, USA.
| |
Collapse
|
38
|
Kohler I, Scherrer AU, Zagordi O, Bianchi M, Wyrzucki A, Steck M, Ledergerber B, Günthard HF, Hangartner L. Prevalence and predictors for homo- and heterosubtypic antibodies against influenza a virus. Clin Infect Dis 2014; 59:1386-93. [PMID: 25139962 DOI: 10.1093/cid/ciu660] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The effectiveness of trivalent influenza vaccination has been confirmed in several studies. To date, it is not known whether repeated exposure and vaccination to influenza promote production of cross-reactive antibodies. Furthermore, how strains encountered earlier in life imprint the immune response is currently poorly understood. METHODS To determine the prevalence for human homo- and heterosubtypic antibody responses, we scrutinized serum samples from 305 healthy volunteers for hemagglutinin-binding and -neutralizing antibodies against several strains and subtypes of influenza A. Statistical analyses were then performed to establish the association of measured values with potential predictors. RESULTS It was found that vaccination not only promoted higher binding and neutralizing antibody titers to homosubtypic influenza isolates but also increased heterosubtypic human immune responses. Both binding and neutralizing antibody titers in relation with age of the donors mirrored the course of the different influenza strain circulation during the last century. Advanced age appeared to be of advantage for both binding and neutralizing titers to most subtypes. In contrast, the first virus subtype encountered was found to imprint to some degree subsequent antibody responses. Antibodies to recent strains, however, primarily seemed to be promoted by vaccination. CONCLUSIONS We provide evidence that vaccinations stimulate both homo- and heterosubtypic immune responses in young and middle-aged as well as more senior individuals. Our analyses suggest that influenza vaccinations not only prevent infection against currently circulating strains but can also stimulate broader humoral immune responses that potentially attenuate infections with zoonotic or antigenically shifted strains.
Collapse
Affiliation(s)
- Ines Kohler
- Institute of Medical Virology, University of Zurich Microbiology and Immunology, Life Science Zurich Graduate School
| | - Alexandra U Scherrer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland
| | | | | | - Arkadiusz Wyrzucki
- Institute of Medical Virology, University of Zurich Microbiology and Immunology, Life Science Zurich Graduate School
| | - Marco Steck
- Institute of Medical Virology, University of Zurich
| | - Bruno Ledergerber
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Switzerland
| | | |
Collapse
|
39
|
Chiu C, Ellebedy AH, Wrammert J, Ahmed R. B cell responses to influenza infection and vaccination. Curr Top Microbiol Immunol 2014; 386:381-98. [PMID: 25193634 DOI: 10.1007/82_2014_425] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Although vaccines against influenza are widely available, control of the disease remains elusive. In part, this is due to the inability of current vaccines to induce durable, broadly protective immune responses. Prevention of influenza depends primarily on effective antibody responses that block virus entry. Following infection, high-affinity IgA antibodies are generated in the respiratory tract that lead to immune exclusion, while IgG prevents systemic spread. These are effective and long-lasting but also exert immune pressure. Mutation of the antigenic determinants of influenza therefore rapidly leads to emergence of novel variants that evade previously generated protective responses. Not only do vaccines suffer from this strain-specific limitation, but also they are suboptimal in their ability to induce durable immunity. However, recent evidence has demonstrated the possibility of inducing broadly cross-reactive antibody responses. Further understanding of the ways in which high-titer, long-lived antibody responses directed against such cross-reactive epitopes can be induced would lead to the development of novel vaccines that may remove the requirement for recurrent vaccination.
Collapse
Affiliation(s)
- Christopher Chiu
- Centre for Respiratory Infection, National Heart and Lung Institute, Imperial College London, London, W2 1PG, UK
| | | | | | | |
Collapse
|
40
|
Gauger PC, Vincent AL. Serum virus neutralization assay for detection and quantitation of serum-neutralizing antibodies to influenza A virus in swine. Methods Mol Biol 2014; 1161:313-24. [PMID: 24899440 DOI: 10.1007/978-1-4939-0758-8_26] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The serum virus neutralization (SVN) assay is a serological test to detect the presence and magnitude of functional systemic antibodies that prevent infectivity of a virus. The SVN assay is a highly sensitive and specific test that may be applied to influenza A viruses in swine to measure the titer of neutralizing antibodies post-infection or after vaccination. Conventional SVN methods performed in vitro are based on inhibition of virus infectivity in cell culture in the presence of neutralizing antibodies. Titer determination may be based on the presence or the absence of cytopathic effect or the evidence of viral infection using an immunoreactive technique. The SVN assay is relatively inexpensive using standard laboratory equipment although it requires cell culture, more time and labor, and technical skill to conduct the assay compared to other serological methods. The SVN test is useful to evaluate the level of serological cross-reactivity between vaccine antisera and variant influenza viruses that may correlate with cross-protection in the host.
Collapse
Affiliation(s)
- Phillip C Gauger
- Veterinary Diagnostic Laboratory, Iowa State University, 1600 South 16th Street, Ames, IA, 50011, USA,
| | | |
Collapse
|
41
|
Donis RO. Antigenic analyses of highly pathogenic avian influenza a viruses. Curr Top Microbiol Immunol 2014; 385:403-40. [PMID: 25190014 DOI: 10.1007/82_2014_422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
In response to the ongoing threat to animal and human health posed by HPAI endemic in poultry, Asia (H5N1) and North America (H7N3) have revived efforts to reduce pandemic risk by disease control at the source and improved pandemic vaccines. Discovery of conserved neutralization epitopes in the HA, which mediate broad protection within and across HA subtypes have changed the paradigm of "broadly reactive" or "universal" vaccine design. Development of such vaccines would benefit from comparative antigenic analysis of viruses with increasing divergence within (and between) HA subtypes. A review of recent work to define the antigenic properties of HPAI viruses revealed data generated through an array of experimental approaches. This information has supported diagnostics and vaccine development for animal and human health. Further harmonization of analytical methods is needed to determine the antigenic relationships among multiple lineages of rapidly evolving HPAI viruses.
Collapse
Affiliation(s)
- Ruben O Donis
- Influenza Division, Centers for Disease Control and Prevention, 1600 Clifton Road NE Mailstop A20, Atlanta, GA, 30333, USA,
| |
Collapse
|
42
|
Serum antibodies against native and denaturated hemagglutinin glycoproteins detected by ELISA as correlates of protection after influenza vaccination in healthy vaccinees and in kidney transplant recipients. J Virol Methods 2013; 193:558-64. [PMID: 23896019 DOI: 10.1016/j.jviromet.2013.07.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 07/15/2013] [Accepted: 07/18/2013] [Indexed: 11/22/2022]
Abstract
The microneutralization assay is the standard method to investigate immune responses to influenza vaccination. However there remains some uncertainty as to whether ELISA results are a true measure of immunity in healthy or immuno-compromised vaccines. Furthermore it has been questioned if antibodies against native ("folded") and against denaturated ("unfolded") viral glycoproteins can equally be used as a marker of protection. In this study, two different quantitative IgG-ELISA assays detecting (i) antibodies against unfolded recombinant hemagglutinin (HA) (r-ELISA) and (ii) antibodies against the native HA on the influenza virus surface captured by fetuin-linkage (f-ELISA) were compared to microneutralization titers in sera from 29 healthy vaccinees (n=87 sera) and 39 kidney transplant recipients (n=117 sera) collected before, three weeks after and six months after vaccination against influenza A (H1N1) 2009. With both ELISAs a significant increase in antibody levels was detected after vaccination and linear regression analysis demonstrated that r-ELISA and f-ELISA correlated with microneutralization (R=0.622 for r-ELISA vs. R=0.56 for f-ELISA). For the healthy vaccinees both ELISAs were found to be adequate to distinguish protected from non-protected individuals (sensitivity and specificity: 87.5%/85.3% for r-ELISA and 87.5%/88.3% for f-ELISA). Results from the transplant recipients showed a slightly reduced sensitivity of 73.3% for r-ELISA while the f-ELISA demonstrated similar sensitivity and specificity as in the healthy vaccinees. However, in order to obtain these assay performances the cut-off-values for protection had to be adjusted for both assays and both investigation cohorts respectively limiting their application in routine laboratories.
Collapse
|
43
|
Improved high-throughput virus neutralisation assay for antibody estimation against pandemic and seasonal influenza strains from 2009 to 2011. J Virol Methods 2013; 189:341-7. [DOI: 10.1016/j.jviromet.2013.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 02/26/2013] [Accepted: 03/05/2013] [Indexed: 11/18/2022]
|
44
|
Development of a neutralization assay for influenza virus using an endpoint assessment based on quantitative reverse-transcription PCR. PLoS One 2013; 8:e56023. [PMID: 23437084 PMCID: PMC3577804 DOI: 10.1371/journal.pone.0056023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 01/09/2013] [Indexed: 12/15/2022] Open
Abstract
A microneutralization assay using an ELISA-based endpoint assessment (ELISA-MN) is widely used to measure the serological response to influenza virus infection and vaccination. We have developed an alternative microneutralization assay for influenza virus using a quantitative reverse transcription PCR-based endpoint assessment (qPCR-MN) in order to improve upon technical limitations associated with ELISA-MN. For qPCR-MN, infected MDCK-London cells in 96-well cell-culture plates are processed with minimal steps such that resulting samples are amenable to high-throughput analysis by downstream one-step quantitative reverse transcription PCR (qRT-PCR; SYBR Green chemistry with primers targeting a conserved region of the M1 gene of influenza A viruses). The growth curves of three recent vaccine strains demonstrated that the qRT-PCR signal detected at 6 hours post-infection reflected an amplification of at least 100-fold over input. Using ferret antisera, we have established the feasibility of measuring virus neutralization at 6 hours post-infection, a duration likely confined to a single virus-replication cycle. The neutralization titer for qPCR-MN was defined as the highest reciprocal serum dilution necessary to achieve a 90% inhibition of the qRT-PCR signal; this endpoint was found to be in agreement with ELISA-MN using the same critical reagents in each assay. qPCR-MN was robust with respect to assay duration (6 hours vs. 12 hours). In addition, qPCR-MN appeared to be compliant with the Percentage Law (i.e., virus neutralization results appear to be consistent over an input virus dose ranging from 500 to 12,000 TCID50). Compared with ELISA-MN, qPCR-MN might have inherent properties conducive to reducing intra- and inter-laboratory variability while affording suitability for automation and high-throughput uses. Finally, our qRT-PCR-based approach may be broadly applicable to the development of neutralization assays for a wide variety of viruses.
Collapse
|
45
|
Mohammed M, Aslan K. Design and Proof-of-Concept Use of a Circular PMMA Platform with 16-Well Sample Capacity for Microwave-Accelerated Bioassays. ACTA ACUST UNITED AC 2013; 5:10-19. [PMID: 24273679 DOI: 10.5101/nbe.v5i1.p20-27] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We demonstrate the design and the proof-of-concept use of a new, circular poly(methyl methacrylate)-based bioassay platform (PMMA platform), which affords for the rapid processing of 16 samples at once. The circular PMMA platform (5 cm in diameter) was coated with a silver nanoparticle film to accelerate the bioassay steps by microwave heating. A model colorimetric bioassay for biotinylated albumin (using streptavidin-labeled horse radish peroxidase) was performed on the PMMA platform coated with and without silver nanoparticles (a control experiment), and at room temperature and using microwave heating. It was shown that the simulated temperature profile of the PMMA platform during microwave heating were comparable to the real-time temperature profile during actual microwave heating of the constructed PMMA platform in a commercial microwave oven. The model colorimetric bioassay for biotinylated albumin was successfully completed in ~2 min (total assay time) using microwave heating, as compared to 90 min at room temperature (total assay time), which indicates a ~45-fold decrease in assay time. Our PMMA platform design afforded for significant reduction in non-specific interactions and low background signal as compared to non-silvered PMMA surfaces when employed in a microwave-accelerated bioassay carried out in a conventional microwave cavity.
Collapse
Affiliation(s)
- Muzaffer Mohammed
- Morgan State University, Department of Chemistry, Baltimore, Maryland 21251
| | | |
Collapse
|
46
|
[Concepts, effectiveness, and perspectives of pandemic and seasonal influenza vaccines]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2013; 56:76-86. [PMID: 23275959 DOI: 10.1007/s00103-012-1590-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
For the first time in history, the conditions to influence the course of an influenza pandemic through vaccination were set during the influenza A H1N1 pandemic in 2009. The specific requirements for pandemic vaccines are to be highly immunogenic in immunologically naive individuals and to be producible quickly in large quantities. In contrast, seasonal influenza vaccines induce a booster response and a broadening of preexisting immunity. In this article the concepts of seasonal and pandemic influenza vaccines and data on their immunogenicity and clinical efficacy are reviewed and discussed. In the upcoming years, seasonal influenza vaccination will continue to be based on inactivated split-virion and subunit vaccines or the live attenuated cold-adapted vaccine. The pandemic vaccines used in 2009 proved to be more immunogenic than expected from prepandemic vaccine trials, while the adverse events observed with AS03-adjuvanted vaccines call their future use into question. However, neither seasonal nor pandemic influenza vaccines can be regarded to be an ideal solution, because they have to be frequently adapted to new virus strains and they lack effectiveness in particular risk groups. They can be regarded as interim approaches to highly immunogenic vaccines that hopefully become available in the future. The underlying principles of future vaccines are also presented in this article.
Collapse
|
47
|
Kucharski AJ, Gog JR. The role of social contacts and original antigenic sin in shaping the age pattern of immunity to seasonal influenza. PLoS Comput Biol 2012; 8:e1002741. [PMID: 23133346 PMCID: PMC3486889 DOI: 10.1371/journal.pcbi.1002741] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/31/2012] [Indexed: 11/23/2022] Open
Abstract
Recent serological studies of seasonal influenza A in humans suggest a striking characteristic profile of immunity against age, which holds across different countries and against different subtypes of influenza. For both H1N1 and H3N2, the proportion of the population seropositive to recently circulated strains peaks in school-age children, reaches a minimum between ages 35–65, then rises again in the older ages. This pattern is little understood. Variable mixing between different age classes can have a profound effect on disease dynamics, and is hence the obvious candidate explanation for the profile, but using a mathematical model of multiple influenza strains, we see that age dependent transmission based on mixing data from social contact surveys cannot on its own explain the observed pattern. Instead, the number of seropositive individuals in a population may be a consequence of ‘original antigenic sin’; if the first infection of a lifetime dominates subsequent immune responses, we demonstrate that it is possible to reproduce the observed relationship between age and seroprevalence. We propose a candidate mechanism for this relationship, by which original antigenic sin, along with antigenic drift and vaccination, results in the age profile of immunity seen in empirical studies. The way in which a population builds immunity to influenza affects outbreak size and the emergence of new strains. However, although age-specific immunity has been widely discussed for the 2009 influenza pandemic, the age profile of immunity to seasonal influenza remains little understood. In contrast to many infections, the proportion of people immune to recent strains peaks in school-age children then reaches a minimum between ages 35–65, before rising again in older age groups. Our results suggest that rather than variable mixing between different age groups being solely responsible, the pattern may be shaped by an effect known as ‘original antigenic sin’, by which the first infection of a lifetime dictates subsequent immune responses: instead of developing antibodies to every new virus that is encountered, the immune system may reuse the response to a similar virus it has already seen. The framework we describe, which extends theoretical models to allow for comparison with data, also opens the possibility of investigating the mechanisms behind patterns of immunity to other evolving pathogens.
Collapse
Affiliation(s)
- Adam J Kucharski
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, United Kingdom.
| | | |
Collapse
|
48
|
Coe CL, Lubach GR, Kinnard J. Immune senescence in old and very old rhesus monkeys: reduced antibody response to influenza vaccination. AGE (DORDRECHT, NETHERLANDS) 2012; 34:1169-77. [PMID: 22231440 PMCID: PMC3448997 DOI: 10.1007/s11357-011-9356-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 12/01/2011] [Indexed: 05/31/2023]
Abstract
The health of old monkeys usually begins to deteriorate by 20 years of age, coinciding with the onset of a slowly progressing immune senescence. Changes in lymphocyte subsets and responses to several antigens have been characterized in geriatric primates, but systematic research has not been conducted on vaccination against influenza virus, a topic of considerable clinical concern for elderly humans. Antibody responses were significantly reduced to primary immunizations in old monkeys, but by administering a second vaccine at 1 month, it was possible to boost antibody titers up to the level found in young adults during their primary phase. The immune competence of unusually long-lived animals (26-37 years) was also compared to more typical aged monkeys (19-25 years). Antibody responses were low overall, although some monkeys in both age groups did respond to immunization. Among the oldest animals, the leukocytes of the responders with higher antibody titers were found to release more interleukin-2 following in vitro stimulation with an anti-CD3/anti-CD28 cocktail relative to their cellular reactions to staphylococcal enterotoxin B. The general decline in immune vigor, and the marked individual variation in how old monkeys age, provides a useful animal model for investigating factors associated with immunosenescence.
Collapse
Affiliation(s)
- Christopher L Coe
- Harlow Center for Biological Psychology, University of Wisconsin, 22 North Charter Street, Madison, WI 53715, USA.
| | | | | |
Collapse
|
49
|
Yang F, He J, Zhong H, Ke C, Zhang X, Hong T, Ni H, Lin J. Temporal trends of influenza A (H1N1) virus seroprevalence following 2009 pandemic wave in Guangdong, China: three cross-sectional serology surveys. PLoS One 2012; 7:e38768. [PMID: 22719938 PMCID: PMC3377711 DOI: 10.1371/journal.pone.0038768] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 05/14/2012] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND To evaluate the temporal trends of seroprevalence to pH1N1 among the Guangdong population following 2009 H1N1 pandemic wave, we conducted three cross-sectional serology surveys in 2010. METHODOLOGY/PRINCIPAL FINDINGS Three surveys were carried out consecutively in 2010 from January 8 to January 24, from March 15 to April 10 and from August 23 to September 4. Sample populations comprising of 4725, 4727, and 4721 subjects respectively were randomly selected for study in these three surveys. The level of antibodies against pH1N1 was evaluated by hemagglutination inhibition assay. In survey 1, the seroprevalence of pH1N1 among all the subjects is 25.1%, declining to 18.4% in survey 2 and increasing to 21.4% in survey 3. Among vaccinated subjects, the seroprevalence was 49.0%, 53.0%, and 49.4% in the three consecutive surveys, showing no significant differences. In contrast, among non-vaccinated subjects, the seroprevalence declined significantly from 22.8% (survey 1) to 14.3% (survey 2) and subsequently increased to 18.1% (survey 3). The multivariate logistic regression analysis revealed that seroprevalence to pH1N1 in non-vaccinated individuals correlated with the investigated order of the surveys, age, and region (all P<0.05). However, it was not correlated with gender (P = 0.650), seasonal influenza vaccination history (P = 0.402) and symptoms (P = 0.074). CONCLUSIONS/SIGNIFICANCE In Guangdong, the seroprevalance to pH1N1 decreased initially and then rebounded modestly during the first 9 months following the 2009 pandemic wave. Our results suggest that the prevalence of pH1N1 is still correlated with age and population density during the post-pandemic period. An early end to the free pH1N1 vaccination program might be another important reason for the slight rebound in seroprevalance. Our study findings can help the Guangdong authorities to make evidence-based decisions about a long-term vaccination strategy and boost immunity in specific population groups (such as children and people living in the capital-city) to prevent further transmission in the future.
Collapse
Affiliation(s)
- Fen Yang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Jianfeng He
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Haojie Zhong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Changwen Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Xin Zhang
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Teng Hong
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Hanzhong Ni
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Jinyan Lin
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou City, Guangdong Province, People’s Republic of China
| |
Collapse
|
50
|
Hansen A, Grund S, Hetzel G, Ivens K, Sümmchen HA, Zgoura P, Hengel H, Adams O, Rump LC. Noncontrolled Trial of Monovalent AS03A-Adjuvanted Vaccine for 2009 Pandemic Influenza A(H1N1) in Long-term Dialysis Patients and Transplant Recipients. Am J Kidney Dis 2012; 59:471-3. [DOI: 10.1053/j.ajkd.2011.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 11/03/2011] [Indexed: 11/11/2022]
|