1
|
Khurana S, King LR, Manischewitz J, Posadas O, Mishra AK, Liu D, Beigel JH, Rappuoli R, Tsang JS, Golding H. Licensed H5N1 vaccines generate cross-neutralizing antibodies against highly pathogenic H5N1 clade 2.3.4.4b influenza virus. Nat Med 2024; 30:2771-2776. [PMID: 39013430 DOI: 10.1038/s41591-024-03189-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/11/2024] [Indexed: 07/18/2024]
Abstract
The emergence of highly pathogenic avian influenza (HPAI) H5N1 clade 2.3.4.4b viruses and their transmission to dairy cattle and animals, including humans, poses a major global public health threat. Therefore, the development of effective vaccines and therapeutics against H5N1 clade 2.3.4.4b virus is considered a public health priority. In the United States, three H5N1 vaccines derived from earlier strains of HPAI H5N1 (A/Vietnam, clade 1, and A/Indonesia, clade 2.1) virus, with (MF59 or AS03) or without adjuvants, are licensed and stockpiled for pre-pandemic preparedness, but whether they can elicit neutralizing antibodies against circulating H5N1 clade 2.3.4.4b viruses is unknown. In this study, we evaluated the binding, hemagglutination inhibition and neutralizing antibody response generated after vaccination of adults with the three licensed vaccines. Individuals vaccinated with the two adjuvanted licensed H5N1 vaccines generated cross-reactive binding and cross-neutralizing antibodies against the HPAI clade 2.3.4.4b A/Astrakhan/3212/2020 virus. Seroconversion rates of 60-95% against H5 clade 2.3.4.4b were observed after two doses of AS03-adjuvanted-A/Indonesia or three doses of MF59-adjuvanted-A/Vietnam vaccine. These findings suggest that the stockpiled US-licensed adjuvanted H5N1 vaccines generate cross-neutralizing antibodies against circulating HPAI H5N1 clade 2.3.4.4b in humans and may be useful as bridging vaccines until updated H5N1 vaccines become available.
Collapse
Affiliation(s)
- Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Silver Spring, MD, USA.
| | - Lisa R King
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Silver Spring, MD, USA
| | - Jody Manischewitz
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Silver Spring, MD, USA
| | - Olivia Posadas
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Silver Spring, MD, USA
| | - Ashish K Mishra
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Silver Spring, MD, USA
| | - Dongxiao Liu
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Silver Spring, MD, USA
| | - John H Beigel
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - John S Tsang
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIH Center for Human Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Departments of Immunobiology and Biomedical Engineering, Yale Center for Systems and Engineering Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
2
|
Wyse CA, Rudderham LM, Nordon EA, Ince LM, Coogan AN, Lopez LM. Circadian Variation in the Response to Vaccination: A Systematic Review and Evidence Appraisal. J Biol Rhythms 2024; 39:219-236. [PMID: 38459699 PMCID: PMC11141079 DOI: 10.1177/07487304241232447] [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] [Indexed: 03/10/2024]
Abstract
Molecular timing mechanisms known as circadian clocks drive endogenous 24-h rhythmicity in most physiological functions, including innate and adaptive immunity. Consequently, the response to immune challenge such as vaccination might depend on the time of day of exposure. This study assessed whether the time of day of vaccination (TODV) is associated with the subsequent immune and clinical response by conducting a systematic review of previous studies. The Cochrane Library, PubMed, Google, Medline, and Embase were searched for studies that reported TODV and immune and clinical outcomes, yielding 3114 studies, 23 of which met the inclusion criteria. The global severe acute respiratory syndrome coronavirus 2 vaccination program facilitated investigation of TODV and almost half of the studies included reported data collected during the COVID-19 pandemic. There was considerable heterogeneity in the demography of participants and type of vaccine, and most studies were biased by failure to account for immune status prior to vaccination, self-selection of vaccination time, or confounding factors such as sleep, chronotype, and shiftwork. The optimum TODV was concluded to be afternoon (5 studies), morning (5 studies), morning and afternoon (1 study), midday (1 study), and morning or late afternoon (1 study), with the remaining 10 studies reporting no effect. Further research is required to understand the relationship between TODV and subsequent immune outcome and whether any clinical benefit outweighs the potential effect of this intervention on vaccine uptake.
Collapse
Affiliation(s)
- Cathy A. Wyse
- Kathleen Lonsdale Institute for Human Health Research and Department of Biology, Maynooth University, Maynooth, Ireland
| | - Laura M. Rudderham
- Kathleen Lonsdale Institute for Human Health Research and Department of Biology, Maynooth University, Maynooth, Ireland
| | - Enya A. Nordon
- Kathleen Lonsdale Institute for Human Health Research and Department of Biology, Maynooth University, Maynooth, Ireland
| | - Louise M. Ince
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Andrew N. Coogan
- Kathleen Lonsdale Institute for Human Health Research and Department of Psychology, Maynooth University, Maynooth, Ireland
| | - Lorna M. Lopez
- Kathleen Lonsdale Institute for Human Health Research and Department of Biology, Maynooth University, Maynooth, Ireland
| |
Collapse
|
3
|
Waldock J, Remarque EJ, Zheng L, Ho S, Hoschler K, Neumann B, Sediri-Schön H, Trombetta CM, Montomoli E, Marchi S, Lapini G, Zhou F, Lartey SL, Cox RJ, Facchini M, Castrucci MR, Friel D, Ollinger T, Caillet C, Music N, Palladino G, Engelhardt OG. Haemagglutination inhibition and virus microneutralisation serology assays: use of harmonised protocols and biological standards in seasonal influenza serology testing and their impact on inter-laboratory variation and assay correlation: A FLUCOP collaborative study. Front Immunol 2023; 14:1155552. [PMID: 37143658 PMCID: PMC10151801 DOI: 10.3389/fimmu.2023.1155552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction The haemagglutination inhibition assay (HAI) and the virus microneutralisation assay (MN) are long-established methods for quantifying antibodies against influenza viruses. Despite their widespread use, both assays require standardisation to improve inter-laboratory agreement in testing. The FLUCOP consortium aims to develop a toolbox of standardised serology assays for seasonal influenza. Building upon previous collaborative studies to harmonise the HAI, in this study the FLUCOP consortium carried out a head-to-head comparison of harmonised HAI and MN protocols to better understand the relationship between HAI and MN titres, and the impact of assay harmonisation and standardisation on inter-laboratory variability and agreement between these methods. Methods In this paper, we present two large international collaborative studies testing harmonised HAI and MN protocols across 10 participating laboratories. In the first, we expanded on previously published work, carrying out HAI testing using egg and cell isolated and propagated wild-type (WT) viruses in addition to high-growth reassortants typically used influenza vaccines strains using HAI. In the second we tested two MN protocols: an overnight ELISA-based format and a 3-5 day format, using reassortant viruses and a WT H3N2 cell isolated virus. As serum panels tested in both studies included many overlapping samples, we were able to look at the correlation of HAI and MN titres across different methods and for different influenza subtypes. Results We showed that the overnight ELISA and 3-5 day MN formats are not comparable, with titre ratios varying across the dynamic range of the assay. However, the ELISA MN and HAI are comparable, and a conversion factor could possibly be calculated. In both studies, the impact of normalising using a study standard was investigated, and we showed that for almost every strain and assay format tested, normalisation significantly reduced inter-laboratory variation, supporting the continued development of antibody standards for seasonal influenza viruses. Normalisation had no impact on the correlation between overnight ELISA and 3-5 day MN formats.
Collapse
Affiliation(s)
- Joanna Waldock
- Influenza Resource Centre, Vaccines, Science Research & Innovation, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Edmond J. Remarque
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Lingyi Zheng
- Department of Research and Development, Sanofi, Marcy L’Etoile, France
| | - Sammy Ho
- Respiratory Viruses Unit, UK Health Secruity Agency, Colindale, United Kingdom
| | - Katja Hoschler
- Respiratory Viruses Unit, UK Health Secruity Agency, Colindale, United Kingdom
| | - Britta Neumann
- Section for Viral Vaccines, Virology Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Hanna Sediri-Schön
- Section for Viral Vaccines, Virology Division, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Claudia M. Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Fan Zhou
- Influenza Centre, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Sarah L. Lartey
- Influenza Centre, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Rebecca J. Cox
- Influenza Centre, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Marzia Facchini
- World Health Organisation (WHO) National Influenza Centre, Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Rita Castrucci
- World Health Organisation (WHO) National Influenza Centre, Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Catherine Caillet
- Department of Research and Development, Sanofi, Marcy L’Etoile, France
| | | | | | - Othmar G. Engelhardt
- Influenza Resource Centre, Vaccines, Science Research & Innovation, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | | |
Collapse
|
4
|
Mangia A, Serra N, Cocomazzi G, Giambra V, Antinucci S, Maiorana A, Giuliani F, Montomoli E, Cantaloni P, Manenti A, Piazzolla V. Cellular and Humoral Immune Responses and Breakthrough Infections After Two Doses of BNT162b Vaccine in Healthcare Workers (HW) 180 Days After the Second Vaccine Dose. Front Public Health 2022; 10:847384. [PMID: 35433614 PMCID: PMC9008351 DOI: 10.3389/fpubh.2022.847384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/21/2022] [Indexed: 01/08/2023] Open
Abstract
Background Immunity and clinical protection induced by mRNA vaccines against SARS-CoV-2 have been shown to decline overtime. To gather information on the immunity profile deemed sufficient in protecting against hospitalization, we tested IgG levels, interferon-gamma (IFN-γ) secretion, and neutralizing antibodies 180 days (d180) after the second shot of BNT162b vaccine, in HW. Methods A total of 392 subjects were enrolled. All received BioNTech/Pfizer from February 2020 to April 2021. The vaccine-specific humoral response was quantitatively determined by testing for IgG anti-S1 domain of SARS-CoV-spike protein. Live virus microneutralization (MN) was evaluated by an assay performing incubation of serial 2-fold dilution of human serum samples, starting from 1:10 to 1:5120, with an equal volume of Wuhan strain and Delta VOC viral solution and assessing the presence/absence of a cytopathic effect. SARS-CoV-2-spike protein-specific T-cell response was determined by a commercial IFN-γ release assay. Results In 352 individuals, at d180, IgG levels decreased substantially but no results below the assay's positivity threshold were observed. Overall, 22 naive (8.1%) had values above the highest threshold. Among COVID-naive, the impact of age, which was observed at earlier stages, disappeared at d180, while it remained significant for 81 who had experienced a previous infection. Following the predictive model of protection by Khoury, we transformed the neutralizing titers in IU/ml and used a 54 IU/ml threshold to identify subjects with 50% protective immunity. Overall, live virus MN showed almost all subjects with previous exposure to SARS-CoV-2 neutralized the virus as compared to 33% of naive double-dosed subjects (p < 0.0001). All previously exposed subjects had strong IFN-γ secretion (>200 mIU/ml); among 271 naive, 7 (2.58%) and 17 (6.27%) subjects did not show borderline or strong secretion, respectively. Conclusions In naive subjects, low IgG titers are relatively long-lasting. Only a third of naive subjects maintain neutralizing responses. After specific stimulation, a very limited number of naive were unable to produce IFN-γ. The results attained in the small group of subjects with breakthrough infection suggest that simultaneous neutralizing antibody titers <20, binding antibody levels/ml <200, and IFN-γ <1,000 mIU/ml in subjects older than 58 may identify at-risk groups.
Collapse
Affiliation(s)
- Alessandra Mangia
- Liver Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Italy
| | - Nicola Serra
- Department of Public Health, University “Federico II”, Naples, Italy
| | - Giovanna Cocomazzi
- Liver Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Italy
| | - Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), San Giovanni Rotondo, Italy
| | - Stefano Antinucci
- Allergy Diagnostic Section Euroimmun, Italy Fondazione “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Italy
| | - Alberto Maiorana
- GSSL Unit, Fondazione “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Italy
| | - Francesco Giuliani
- ICT Innovation and Research Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi Srl, Siena, Italy
| | | | | | - Valeria Piazzolla
- Liver Unit, Fondazione IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Italy
| |
Collapse
|
5
|
Manenti A, Molesti E, Maggetti M, Torelli A, Lapini G, Montomoli E. The theory and practice of the viral dose in neutralization assay: Insights on SARS-CoV-2 "doublethink" effect. J Virol Methods 2021; 297:114261. [PMID: 34403775 PMCID: PMC8364219 DOI: 10.1016/j.jviromet.2021.114261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 11/26/2022]
Abstract
The neutralization assays are considered the gold-standard being capable of evaluating and detecting, functional antibodies. To date, many different protocols exist for micro-neutralization (MN) assay which varies in several steps: cell number and seeding conditions, virus amount used in the infection step, virus-serum-cells incubation time and read out. The aim of the present preliminary study was to carry out SARS-CoV-2 wild type MN assay in order to investigate which optimal tissue culture infective dose 50 (TCID50) infective dose in use is the most adequate choice for implementation in terms of reproducibility, standardization possibilities and comparability of results. Therefore, we assessed the MN by using two viral infective doses: the “standard” dose of 100 TCID50/well and a reduced dose of 25 TCID50/well. The results obtained, yielded by MN on using the lower infective dose (25 TCID50), were higher respect to those obtained with the standard infective dose. This suggests that the lower dose can potentially have a positive impact on the detection and estimation of real amount of neutralizing antibodies present in a given sample, showing higher sensitivity maintaining high specificity.
Collapse
Affiliation(s)
| | | | | | | | | | - Emanuele Montomoli
- VisMederi s.r.l., Siena, Italy; VisMederi Research s.r.l., Siena, Italy; Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
6
|
Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med 2021; 27:1205-1211. [PMID: 34002089 DOI: 10.1038/s41591-021-01377-8] [Citation(s) in RCA: 2550] [Impact Index Per Article: 850.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
Predictive models of immune protection from COVID-19 are urgently needed to identify correlates of protection to assist in the future deployment of vaccines. To address this, we analyzed the relationship between in vitro neutralization levels and the observed protection from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection using data from seven current vaccines and from convalescent cohorts. We estimated the neutralization level for 50% protection against detectable SARS-CoV-2 infection to be 20.2% of the mean convalescent level (95% confidence interval (CI) = 14.4-28.4%). The estimated neutralization level required for 50% protection from severe infection was significantly lower (3% of the mean convalescent level; 95% CI = 0.7-13%, P = 0.0004). Modeling of the decay of the neutralization titer over the first 250 d after immunization predicts that a significant loss in protection from SARS-CoV-2 infection will occur, although protection from severe disease should be largely retained. Neutralization titers against some SARS-CoV-2 variants of concern are reduced compared with the vaccine strain, and our model predicts the relationship between neutralization and efficacy against viral variants. Here, we show that neutralization level is highly predictive of immune protection, and provide an evidence-based model of SARS-CoV-2 immune protection that will assist in developing vaccine strategies to control the future trajectory of the pandemic.
Collapse
|
7
|
Creanga A, Gillespie RA, Fisher BE, Andrews SF, Lederhofer J, Yap C, Hatch L, Stephens T, Tsybovsky Y, Crank MC, Ledgerwood JE, McDermott AB, Mascola JR, Graham BS, Kanekiyo M. A comprehensive influenza reporter virus panel for high-throughput deep profiling of neutralizing antibodies. Nat Commun 2021; 12:1722. [PMID: 33741916 PMCID: PMC7979723 DOI: 10.1038/s41467-021-21954-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/22/2021] [Indexed: 01/31/2023] Open
Abstract
Broadly neutralizing antibodies (bnAbs) have been developed as potential countermeasures for seasonal and pandemic influenza. Deep characterization of these bnAbs and polyclonal sera provides pivotal understanding for influenza immunity and informs effective vaccine design. However, conventional virus neutralization assays require high-containment laboratories and are difficult to standardize and roboticize. Here, we build a panel of engineered influenza viruses carrying a reporter gene to replace an essential viral gene, and develop an assay using the panel for in-depth profiling of neutralizing antibodies. Replication of these viruses is restricted to cells expressing the missing viral gene, allowing it to be manipulated in a biosafety level 2 environment. We generate the neutralization profile of 24 bnAbs using a 55-virus panel encompassing the near-complete diversity of human H1N1 and H3N2, as well as pandemic subtype viruses. Our system offers in-depth profiling of influenza immunity, including the antibodies against the hemagglutinin stem, a major target of universal influenza vaccines.
Collapse
Affiliation(s)
- Adrian Creanga
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca A Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brian E Fisher
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah F Andrews
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julia Lederhofer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christina Yap
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Liam Hatch
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tyler Stephens
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Michelle C Crank
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Chu DKW, Perera RAPM, Ali A, Oladipo JO, Mamo G, So RTY, Zhou Z, Chor YY, Chan CK, Belay D, Tayachew A, Mengesha M, Regassa F, Lam NT, Poon LLM, Peiris M. Influenza A Virus Infections in Dromedary Camels, Nigeria and Ethiopia, 2015-2017. Emerg Infect Dis 2021; 26:173-176. [PMID: 31855544 PMCID: PMC6924894 DOI: 10.3201/eid2601.191165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We examined nasal swabs and serum samples acquired from dromedary camels in Nigeria and Ethiopia during 2015–2017 for evidence of influenza virus infection. We detected antibodies against influenza A(H1N1) and A(H3N2) viruses and isolated an influenza A(H1N1)pdm09–like virus from a camel in Nigeria. Influenza surveillance in dromedary camels is needed.
Collapse
|
9
|
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: 0.3] [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
|
10
|
Kim KH, Jung YJ, Lee Y, Park BR, Oh J, Lee YN, Kim MC, Jeeva S, Kang SM. Cross protection by inactivated recombinant influenza viruses containing chimeric hemagglutinin conjugates with a conserved neuraminidase or M2 ectodomain epitope. Virology 2020; 550:51-60. [PMID: 32882637 DOI: 10.1016/j.virol.2020.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
Influenza virus neuraminidase (NA) contains a universally conserved epitope (NAe, NA222-230). However, no studies have reported vaccines targeting this NA conserved epitope and inducing antibodies recognizing NAe. The extracellular domain of M2 (M2e) is considered as an attractive target for a universal influenza vaccine. We generated recombinant influenza H1N1 viruses expressing conserved epitopes in hemagglutinin (HA) molecules: NAe (NAe-HA) or M2e (M2e-HA) within the HA head domain. Inactivated recombinant NAe-HA and M2e-HA viruses were more effective in inducing IgG antibodies specific for an inserted conserved epitope than live recombinant virus. Recombinant inactivated M2e-HA virus vaccination induced cross protection against H3N2 virus with less weight loss compared to NAe-HA and was more effective in inducing humoral and cellular M2e immune responses. This study provides insight into developing recombinant influenza virus vaccines compatible with current platforms to induce antibody responses to conserved poorly immunogenic epitopes.
Collapse
Affiliation(s)
- Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Youri Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Bo Ryoung Park
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Judy Oh
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yu-Na Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; Animal and Plant Quarantine Agency, Gimcheon, 39660, Republic of Korea
| | - Min-Chul Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; CARESIDE Co., Ltd., Seongnam, Gyeonggi-do, Republic of Korea
| | - Subbiah Jeeva
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
11
|
Hay JA, Minter A, Ainslie KEC, Lessler J, Yang B, Cummings DAT, Kucharski AJ, Riley S. An open source tool to infer epidemiological and immunological dynamics from serological data: serosolver. PLoS Comput Biol 2020; 16:e1007840. [PMID: 32365062 PMCID: PMC7241836 DOI: 10.1371/journal.pcbi.1007840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/21/2020] [Accepted: 04/01/2020] [Indexed: 11/19/2022] Open
Abstract
We present a flexible, open source R package designed to obtain biological and epidemiological insights from serological datasets. Characterising past exposures for multi-strain pathogens poses a specific statistical challenge: observed antibody responses measured in serological assays depend on multiple unobserved prior infections that produce cross-reactive antibody responses. We provide a general modelling framework to jointly infer infection histories and describe immune responses generated by these infections using antibody titres against current and historical strains. We do this by linking latent infection dynamics with a mechanistic model of antibody kinetics that generates expected antibody titres over time. Our aim is to provide a flexible package to identify infection histories that can be applied to a range of pathogens. We present two case studies to illustrate how our model can infer key immunological parameters, such as antibody titre boosting, waning and cross-reaction, as well as latent epidemiological processes such as attack rates and age-stratified infection risk.
Collapse
Affiliation(s)
- James A. Hay
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Amanda Minter
- Centre for the Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Kylie E. C. Ainslie
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| | - Justin Lessler
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Bingyi Yang
- Department of Biology, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Derek A. T. Cummings
- Department of Biology, University of Florida, Gainesville, Florida, United States of America
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Adam J. Kucharski
- Centre for the Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Steven Riley
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
12
|
Shi WZ, Jiang LZ, Song GP, Wang S, Xiong P, Ke CW. Study on the Antiviral Activities and Hemagglutinin-Based Molecular Mechanism of Novel Chlorogenin 3- O-β-Chacotrioside Derivatives Against H5N1 Subtype Viruses. Viruses 2020; 12:E304. [PMID: 32168921 PMCID: PMC7150989 DOI: 10.3390/v12030304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/04/2020] [Accepted: 03/07/2020] [Indexed: 12/27/2022] Open
Abstract
The objective of this study was to investigate the inhibitory effect of chlorogenin 3-O-β-chacotrioside derivatives against H5N1 subtype of the highly pathogenic avian influenza (HPAI) viruses and its molecular mechanism. A series of novel small molecule pentacyclic triterpene derivatives were designed and synthesized and their antiviral activities on HPAI H5N1 viruses were detected. The results displayed that the derivatives UA-Nu-ph-5, XC-27-1 and XC-27-2 strongly inhibited wild-type A/Duck/Guangdong/212/2004 H5N1 viruses with the IC50 values of 15.59 ± 2.4 μM, 16.83 ± 1.45 μM, and 12.45 ± 2.27 μM, respectively, and had the selectivity index (SI) > 3, which was consistent with the efficacy against A/Thailand/kan353/2004 pseudo-typed viruses. Four dealt patterns were compared via PRNT. The prevention dealt pattern showed the strongest inhibitory effects than other patterns, suggesting that these derivatives act on the entry process at the early stages of H5N1 viral infection, providing protection for cells against infection. Further studies through hemagglutinin inhibition (HI) and neuraminidase inhibitory (NAI) assay confirmed that these derivatives inhibited H5N1 virus replication by interfering with the viral hemagglutinin function. The derivatives could recognize specifically HA protein with binding affinity constant KD values of 2.57 × 10-4 M and 3.67 × 10-4 M. In addition, through site-directed mutagenesis combined with a pseudovirion system, we identified that the high-affinity docking sites underlying interaction were closely associated with amino acid residues I391 and T395 of HA. However, the potential binding sites of the derivatives with HA did not locate at HA1 sialic acids receptor binding domain (RBD). Taken together, these study data manifested that chlorogenin 3-O-β-chacotrioside derivatives generated antiviral effect against HPAI H5N1 viruses by targeting the hemagglutinin fusion machinery.
Collapse
Affiliation(s)
- Wan-Zhen Shi
- Department of Pharmaceutical Engineering, South China Agricultural University, Guangzhou 510640, China; (W.-Z.S.); (G.-P.S.)
| | - Ling-Zhi Jiang
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China;
| | - Gao-Peng Song
- Department of Pharmaceutical Engineering, South China Agricultural University, Guangzhou 510640, China; (W.-Z.S.); (G.-P.S.)
| | - Sheng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Ping Xiong
- Department of Pharmaceutical Engineering, South China Agricultural University, Guangzhou 510640, China; (W.-Z.S.); (G.-P.S.)
| | - Chang-Wen Ke
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, China;
| |
Collapse
|
13
|
Comparison of influenza-specific neutralizing antibody titers determined using different assay readouts and hemagglutination inhibition titers: good correlation but poor agreement. Vaccine 2020; 38:2527-2541. [PMID: 32044163 DOI: 10.1016/j.vaccine.2020.01.088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/23/2020] [Accepted: 01/29/2020] [Indexed: 12/23/2022]
Abstract
Determination of influenza-specific antibody titers is commonly done using the hemagglutination inhibition assay (HAI) and the viral microneutralization assay (MN). Both assays are characterized by high intra- and inter-laboratory variability. The HAI assay offers little opportunity for standardization. For the MN assay, variability might be due to the use of different assay protocols employing different readouts. We therefore aimed at investigating which of the MN assay readout methods currently in use would be the most suitable choice for a standardized MN assay that could serve as a substitute for the HAI assay. For this purpose, human serum samples were tested for the presence of influenza specific neutralizing antibodies against A/California/7/09 H1N1 (49 sera) or A/Hong Kong/4801/2014 (50 sera) using four different infection readout methods for the MN assay (cytopathic effect, hemagglutination, ELISA, RT qPCR) and using the HAI assay. The results were compared by correlation analysis and by determining the level of agreement before and after normalization to a standard serum. Titers as measured by the 4 MN assay readouts showed good correlation, with high Person's r for most comparisons. However, agreement between nominal titers varied with readouts compared and virus strain used. In addition, Pearson's correlation of MN titers with HAI titers was high but agreement of nominal titers was moderate and the average difference between the readings of two assays (bias) was virus strain-dependent. Normalization to a standard serum did not result in better agreement of assay results. Our study demonstrates that different MN readouts result in nominally different antibody titers. Accordingly, the use of a common and standardized MN assay protocol will be crucial to minimize inter-laboratory variability. Based on reproducibility, cost effectiveness and unbiased assessment of results we elected the MN assay with ELISA readout as most suitable for a possible replacement of the HAI assay.
Collapse
|
14
|
Portnoff AD, Patel N, Massare MJ, Zhou H, Tian JH, Zhou B, Shinde V, Glenn GM, Smith G. Influenza Hemagglutinin Nanoparticle Vaccine Elicits Broadly Neutralizing Antibodies against Structurally Distinct Domains of H3N2 HA. Vaccines (Basel) 2020; 8:vaccines8010099. [PMID: 32098409 PMCID: PMC7157642 DOI: 10.3390/vaccines8010099] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 11/30/2022] Open
Abstract
Influenza vaccine effectiveness varies annually due to the fast evolving seasonal influenza A(H3N2) strain and egg-derived mutations—both of which can cause a mismatch between the vaccine and circulating strains. To address these limitations, we have developed a hemagglutinin (HA)-based protein-detergent nanoparticle influenza vaccine (NIV) with a saponin-based Matrix-M™ adjuvant. In a phase 1 clinical trial of older adults, the vaccine demonstrated broadly cross-reactive A(H3N2) HA antibody responses. Two broadly neutralizing monoclonal antibodies derived from NIV-immunized mice were characterized by transmission electron microscopy (TEM), antibody competition assays, fluorescence-activated cell sorting (FACS) analysis, and protein–protein docking. These antibodies recognize two conserved regions of the head domain, namely the receptor binding site and the vestigial esterase subdomain, thus demonstrating the potential for an HA subunit vaccine to elicit antibodies targeting structurally and antigenically distinct but conserved sites. Antibody competition studies with sera from the phase 1 trial in older adults confirmed that humans also make antibodies to these two head domains and against the highly conserved stem domain. This data supports the potential of an adjuvanted recombinant HA nanoparticle vaccine to induce broadly protective immunity and improved vaccine efficacy.
Collapse
|
15
|
An efficient, reproducible and accurate RT-qPCR based method to determine mumps specific neutralizing antibody. J Virol Methods 2020; 277:113817. [PMID: 31911119 DOI: 10.1016/j.jviromet.2020.113817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 11/29/2019] [Accepted: 01/03/2020] [Indexed: 11/22/2022]
Abstract
INTRODUCTION A resurgence of mumps among fully vaccinated adolescents and young adults globally has led to questions about the longevity of vaccine derived specific immunity. Unfortunately, the ideal serological correlate of immunity to mumps has yet to be identified. However, neutralising antibody titres in serum are used extensively as a surrogate marker of immunity to mumps. Conventional neutralisation tests are technically challenging, thus we developed and validated a high throughput, RT-qPCR microneutralisation (RT-qPCR-MN) method to determine serum neutralising antibody levels to mumps virus strains which avoids a number of the technical limitations of existing methods. METHODS The qPCR-MN assays were thoroughly validated using human serum samples from patients with prior exposure to mumps infection or vaccination. RESULTS Each sample of pooled sera neutralised virus at a constant rate and without significant changes when tested against genotype A (MuV-A) and G (MuV-G) mumps virus concentrations from 200 to 3200 TCID50. The within run and between run variation of the RT-qPCR-MN assays for both genotypes were less than 3 % and 9 % for low and high titre samples, respectively. The correlation between the focus reduction neutralisation test and RT-qPCR-MN was excellent for both MuV-G (r2 = 0.80, 95CI: 0.67-1.00, p < 0.0001) and MuV-A genotypes (r2 = 0.88, 95 %CI 0.69-1.00, p < 0.0001) endpoint determinations. CONCLUSIONS We have developed a RT-qPCR MN assay for mumps virus that is simple, fast, scientifically objective and has high throughput. The assay can be used to provide key insights into the efficacy of mumps vaccination, to help explain the causes for the resurgence of mumps infection in vaccinated populations.
Collapse
|
16
|
Krammer F, Weir JP, Engelhardt O, Katz JM, Cox RJ. Meeting report and review: Immunological assays and correlates of protection for next-generation influenza vaccines. Influenza Other Respir Viruses 2019; 14:237-243. [PMID: 31837101 PMCID: PMC7040967 DOI: 10.1111/irv.12706] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND This report summarizes the discussions and conclusions from the "Immunological Assays and Correlates of Protection for Next-Generation Influenza Vaccines" meeting which took place in Siena, Italy, from March 31, 2019, to April 2, 2019. CONCLUSIONS Furthermore, we review current correlates of protection against influenza virus infection and disease and their usefulness for the development of next generation broadly protective and universal influenza virus vaccines.
Collapse
Affiliation(s)
- Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Jerry P Weir
- Division of Viral Products, Food and Drug Administration, Bethesda, MD, USA
| | - Othmar Engelhardt
- Division of Virology, National Institute for Biological Standards and Control, Potters Bar, UK
| | - Jacqueline M Katz
- Formerly Influenza Branch, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Rebecca Jane Cox
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway
| |
Collapse
|
17
|
Assessment of population susceptibility to upcoming seasonal influenza epidemic strain using interepidemic emerging influenza virus strains. Epidemiol Infect 2019; 147:e279. [PMID: 31556360 PMCID: PMC6805736 DOI: 10.1017/s0950268819001717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Seasonal influenza virus epidemics have a major impact on healthcare systems. Data on population susceptibility to emerging influenza virus strains during the interepidemic period can guide planning for resource allocation of an upcoming influenza season. This study sought to assess the population susceptibility to representative emerging influenza virus strains collected during the interepidemic period. The microneutralisation antibody titers (MN titers) of a human serum panel against representative emerging influenza strains collected during the interepidemic period before the 2018/2019 winter influenza season (H1N1-inter and H3N2-inter) were compared with those against influenza strains representative of previous epidemics (H1N1-pre and H3N2-pre). A multifaceted approach, incorporating both genetic and antigenic data, was used in selecting these representative influenza virus strains for the MN assay. A significantly higher proportion of individuals had a ⩾four-fold reduction in MN titers between H1N1-inter and H1N1-pre than that between H3N2-inter and H3N2-pre (28.5% (127/445) vs. 4.9% (22/445), P < 0.001). The geometric mean titer (GMT) of H1N1-inter was significantly lower than that of H1N1-pre (381 (95% CI 339-428) vs. 713 (95% CI 641-792), P < 0.001), while there was no significant difference in the GMT between H3N2-inter and H3N2-pre. Since A(H1N1) predominated the 2018-2019 winter influenza epidemic, our results corroborated the epidemic subtype.
Collapse
|
18
|
Biuso F, Palladino L, Manenti A, Stanzani V, Lapini G, Gao J, Couzens L, Eichelberger MC, Montomoli E. Use of lentiviral pseudotypes as an alternative to reassortant or Triton X-100-treated wild-type Influenza viruses in the neuraminidase inhibition enzyme-linked lectin assay. Influenza Other Respir Viruses 2019; 13:504-516. [PMID: 31411006 PMCID: PMC6692537 DOI: 10.1111/irv.12669] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Formulation of neuraminidase (NA) within influenza vaccines is gaining importance in light of recent human studies. The enzyme-linked lectin assay (ELLA) is considered a reliable assay to evaluate human anti-NA antibodies. OBJECTIVES To overcome interference by hemagglutinin (HA)-specific antibodies and detect neuraminidase inhibitory (NI) antibodies only, two different sources of antigen have been studied in ELLA: reassortant viruses with a mismatched avian origin-HA or Triton X-100 (Tx)-treated wild-type viruses. Pseudotypes or pseudovirus (PV), characterized by a lentivirus core bearing human influenza NA and avian influenza HA, were investigated as an alternative source of antigen and compared to HA-mismatched and Tx-treated viruses, since represent a safer product to be handled. METHODS Two independent panels of sera were analyzed by ELLA to evaluate the anti-NA response against N1 (A/California/07/2009 (H1N1pdm)) and N2 (A/Hong Kong/4801/2014 (H3N2)). The NA inhibition (NI) antibody titers measured as either the 50% end point or 50% inhibitory concentration (IC50 ) were compared for every source of antigen. RESULTS The ELLA assay performed well with all three sources of antigen. NI titers measured using each antigen type correlated well when reported either as end point titers or as the IC50 . CONCLUSIONS This study suggests that HA-mismatched whole virus, Triton-treated wild-type virus or PV can be used to measure NI antibody titers of human sera, but further comparability/validation assays should be performed to assess statistical differences. The data support the use of PV as an attractive alternative source of antigen and justify further investigation to improve stability of this antigen source.
Collapse
Affiliation(s)
- Fabrizio Biuso
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- VisMederi Research s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | - Laura Palladino
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | - Alessandro Manenti
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- VisMederi Research s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | | | - Giulia Lapini
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
| | - Jin Gao
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMDUSA
| | - Laura Couzens
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMDUSA
| | - Maryna C. Eichelberger
- Center for Biologics Evaluation and Research, Food and Drug AdministrationSilver SpringMDUSA
| | - Emanuele Montomoli
- VisMederi s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- VisMederi Research s.r.l., Strada del Petriccio e BelriguardoSienaItaly
- Department of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| |
Collapse
|
19
|
Abstract
Clinical trials of vaccines differ from those of drugs both practically and ethically. Influenza vaccine trials pose additional challenges which flow from the seasonality of the disease and vaccine programmes and the changeability of circulating strains and epidemic size. Serological correlates are widely used in evaluating influenza vaccines but interpreting such responses is also difficult. Development and testing of vaccines for deployment in an influenza pandemic combines all these obstacles with extreme unpredictability, enormous urgency and the need for very large numbers of doses. Advances in manufacturing technology, diagnostics, identification of stable protective antigens and epitopes, new adjuvants and improved understanding of indirect and downstream effects may help to alleviate these difficulties in the future.
Collapse
|
20
|
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: 0.4] [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
|
21
|
Zhao H, Xu K, Jiang Z, Shao M, Liu S, Li X, Wang J, Li C. A neuraminidase activity-based microneutralization assay for evaluating antibody responses to influenza H5 and H7 vaccines. PLoS One 2018; 13:e0207431. [PMID: 30440054 PMCID: PMC6237356 DOI: 10.1371/journal.pone.0207431] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 10/31/2018] [Indexed: 11/20/2022] Open
Abstract
Outbreaks of the highly pathogenic avian influenza H5N1 and H7N9 viruses have spurred an unprecedented research effort to develop antivirals and vaccines against influenza. Standardized methods for vaccine evaluation are critical for facilitating vaccine development. Compared with hemagglutination inhibition assays, mounting evidence suggest that microneutralization tests (MNTs) is a better choice for the evaluation of candidate pandemic influenza vaccines because they measure neutralizing antibody activity in cell cultures and are more sensitive in detecting H5 and H7. Here, we report a MNT measuring neuraminidase activity as the read-out (NA-MNT) for quantitative analysis of neutralizing antibodies against avian influenza viruses. Compared to the conventional microneutralization assay (ELISA-MNT), the NA-MNT is faster with lower intra- and inter-assay variations, while no difference in geometric mean titers was found between these two assays for the evaluation of H5N1 and H7N9 vaccines. These results suggest that NA-MNT is a reliable and high throughput method which could facilitate the development of candidate pandemic influenza vaccine.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antibody Formation/immunology
- Birds/virology
- Hemagglutinin Glycoproteins, Influenza Virus
- Humans
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza A Virus, H7N9 Subtype/pathogenicity
- Influenza Vaccines/immunology
- Influenza in Birds/immunology
- Influenza in Birds/virology
- Influenza, Human/genetics
- Influenza, Human/immunology
- Influenza, Human/virology
- Neuraminidase/genetics
- Neuraminidase/immunology
- Pandemics
- Vaccines, Inactivated/immunology
Collapse
Affiliation(s)
- Hui Zhao
- Division of Viral Vaccine III, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, P.R. China
| | - Kangwei Xu
- Division of Viral Vaccine III, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, P.R. China
| | - Zheng Jiang
- Division of Viral Vaccine III, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, P.R. China
| | - Ming Shao
- Division of Viral Vaccine III, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, P.R. China
| | - Shuzhen Liu
- Division of Viral Vaccine III, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, P.R. China
| | - Xuguang Li
- Center for Biologics Evaluation, Ottawa, Biologics and Genetic Therapies Directorate, Health Canada and WHO Collaborating Center for Standardization and Evaluation, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Junzhi Wang
- Division of Viral Vaccine III, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, P.R. China
- * E-mail: (JW); (CL)
| | - Changgui Li
- Division of Viral Vaccine III, National Institutes for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, P.R. China
- * E-mail: (JW); (CL)
| |
Collapse
|
22
|
Agarwal D, Schmader KE, Kossenkov AV, Doyle S, Kurupati R, Ertl HCJ. Immune response to influenza vaccination in the elderly is altered by chronic medication use. Immun Ageing 2018; 15:19. [PMID: 30186359 PMCID: PMC6119322 DOI: 10.1186/s12979-018-0124-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/21/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The elderly patient population is the most susceptible to influenza virus infection and its associated complications. Polypharmacy is common in the aged, who often have multiple co-morbidities. Previous studies have demonstrated that commonly used prescription drugs can have extensive impact on immune defenses and responses to vaccination. In this study, we examined how the dynamics of immune responses to the two influenza A virus strains of the trivalent inactivated influenza vaccine (TIV) can be affected by patient's history of using the prescription drugs Metformin, NSAIDs or Statins. RESULTS We provide evidence for differential antibody (Ab) production, B-cell phenotypic changes, alteration in immune cell proportions and transcriptome-wide perturbation in individuals with a history of long-term medication use, compared with non-users. We noted a diminished response to TIV in the elderly on Metformin, whereas those on NSAIDs or Statins had higher baseline responses but reduced relative increases in virus-neutralizing Abs (VNAs) or Abs detected by an enzyme-linked immunosorbent assay (ELISA) following vaccination. CONCLUSION Collectively, our findings suggest novel pathways that might underlie how long-term medication use impacts immune response to influenza vaccination in the elderly. They provide a strong rationale for targeting the medication-immunity interaction in the aged population to improve vaccination responses.
Collapse
Affiliation(s)
- Divyansh Agarwal
- Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Kenneth E. Schmader
- Division of Geriatrics, Duke University Medical Center; Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC 27705 USA
| | | | - Susan Doyle
- Division of Geriatrics, Duke University Medical Center; Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC 27705 USA
| | - Raj Kurupati
- The Wistar Institute, Philadelphia, PA 19104 USA
| | | |
Collapse
|
23
|
Makarkov AI, Patel AR, Bainov V, Ward BJ. A novel serological assay for influenza based on DiD fluorescence dequenching that is free from observer bias and potentially automatable – A proof of concept study. Vaccine 2018; 36:4485-4493. [DOI: 10.1016/j.vaccine.2018.06.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/06/2018] [Accepted: 06/08/2018] [Indexed: 10/28/2022]
|
24
|
Trost JF, LeMasters EH, Liu F, Carney P, Lu X, Sugawara K, Hongo S, Stevens J, Steinhauer DA, Tumpey T, Katz JM, Levine MZ, Li ZN. Development of a high-throughput assay to detect antibody inhibition of low pH induced conformational changes of influenza virus hemagglutinin. PLoS One 2018; 13:e0199683. [PMID: 29949635 PMCID: PMC6021090 DOI: 10.1371/journal.pone.0199683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/12/2018] [Indexed: 12/14/2022] Open
Abstract
Many broadly neutralizing antibodies (bnAbs) bind to conserved areas of the hemagglutinin (HA) stalk region and can inhibit the low pH induced HA conformational changes necessary for viral membrane fusion activity. We developed and evaluated a high-throughput virus-free and cell-free ELISA based low pH induced HA Conformational Change Inhibition Antibody Detection Assay (HCCIA) and a complementary proteinase susceptibility assay. Human serum samples (n = 150) were tested by HCCIA using H3 recombinant HA. Optical density (OD) ratios of mAb HC31 at pH 4.8 to pH 7.0 ranged from 0.87 to 0.09. Our results demonstrated that low pH induced HA conformational change inhibition antibodies (CCI) neutralized multiple H3 strains after removal of head-binding antibodies. The results suggest that HCCIA can be utilized to detect and characterize CCI in sera, that are potentially broadly neutralizing, and serves as a useful tool for evaluating universal vaccine candidates targeting the HA stalk.
Collapse
MESH Headings
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Enzyme-Linked Immunosorbent Assay/methods
- Hemagglutinin Glycoproteins, Influenza Virus/blood
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- High-Throughput Screening Assays/methods
- Humans
- Hydrogen-Ion Concentration
- Influenza A Virus, H3N2 Subtype/chemistry
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza, Human/blood
- Influenza, Human/immunology
- Models, Molecular
- Protein Conformation
- Recombinant Proteins/chemistry
- Recombinant Proteins/immunology
Collapse
Affiliation(s)
- Jessica F. Trost
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Elizabeth H. LeMasters
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Feng Liu
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Paul Carney
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Xiuhua Lu
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Kanetsu Sugawara
- Department of Infectious Diseases, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Seiji Hongo
- Department of Infectious Diseases, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - James Stevens
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - David A. Steinhauer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Terrence Tumpey
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jacqueline M. Katz
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Min Z. Levine
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Zhu-Nan Li
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
25
|
Wang J, Hilchey SP, DeDiego M, Perry S, Hyrien O, Nogales A, Garigen J, Amanat F, Huertas N, Krammer F, Martinez-Sobrido L, Topham DJ, Treanor JJ, Sangster MY, Zand MS. Broad cross-reactive IgG responses elicited by adjuvanted vaccination with recombinant influenza hemagglutinin (rHA) in ferrets and mice. PLoS One 2018; 13:e0193680. [PMID: 29641537 PMCID: PMC5894995 DOI: 10.1371/journal.pone.0193680] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/15/2018] [Indexed: 12/17/2022] Open
Abstract
Annual immunization against influenza virus is a large international public health effort. Accumulating evidence suggests that antibody mediated cross-reactive immunity against influenza hemagglutinin (HA) strongly correlates with long-lasting cross-protection against influenza virus strains that differ from the primary infection or vaccination strain. However, the optimal strategies for achieving highly cross-reactive antibodies to the influenza virus HA have not yet to be defined. In the current study, using Luminex-based mPlex-Flu assay, developed by our laboratory, to quantitatively measure influenza specific IgG antibody mediated cross-reactivity, we found that prime-boost-boost vaccination of ferrets with rHA proteins admixed with adjuvant elicited higher magnitude and broader cross-reactive antibody responses than that induced by actual influenza viral infection, and this cross-reactive response likely correlated with increased anti-stalk reactive antibodies. We observed a similar phenomenon in mice receiving three sequential vaccinations with rHA proteins from either A/California/07/2009 (H1N1) or A/Hong Kong/1/1968 (H3N2) viruses admixed with Addavax, an MF59-like adjuvant. Using this same mouse vaccination model, we determined that Addavax plays a more significant role in the initial priming event than in subsequent boosts. We also characterized the generation of cross-reactive antibody secreting cells (ASCs) and memory B cells (MBCs) when comparing vaccination to viral infection. We have also found that adjuvant plays a critical role in the generation of long-lived ASCs and MBCs cross-reactive to influenza viruses as a result of vaccination with rHA of influenza virus, and the observed increase in stalk-reactive antibodies likely contributes to this IgG mediated broad cross-reactivity.
Collapse
Affiliation(s)
- Jiong Wang
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Shannon P. Hilchey
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Marta DeDiego
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Sheldon Perry
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Ollivier Hyrien
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Aitor Nogales
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jessica Garigen
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Fatima Amanat
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Nelson Huertas
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Luis Martinez-Sobrido
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - David J. Topham
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - John J. Treanor
- Division of Infectious Disease, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Mark Y. Sangster
- Biostatistics, Bioinformatics, and Epidemiology Program, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Martin S. Zand
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States of America
- Rochester Center for Health Informatics, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
26
|
Crowe JE. Principles of Broad and Potent Antiviral Human Antibodies: Insights for Vaccine Design. Cell Host Microbe 2018; 22:193-206. [PMID: 28799905 DOI: 10.1016/j.chom.2017.07.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Antibodies are the principal immune effectors that mediate protection against reinfection following viral infection or vaccination. Robust techniques for human mAb isolation have been developed in the last decade. The study of human mAbs isolated from subjects with prior immunity has become a mainstay for rational structure-based, next-generation vaccine development. The plethora of detailed molecular and genetic studies coupling the structure of antigen-antibody complexes with their antiviral function has begun to reveal common principles of critical interactions on which we can build better vaccines and therapeutic antibodies. This review outlines the approaches to isolating and studying human antiviral mAbs and discusses the common principles underlying the basis for their activity. This review also examines progress toward the goal of achieving a comprehensive understanding of the chemical and physical basis for molecular recognition of viral surface proteins in order to build predictive molecular models that can be used for vaccine design.
Collapse
Affiliation(s)
- James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
27
|
Zhu H, Lee ACY, Li C, Mak WWN, Chen YY, Chan KH, Zhang AJX, Fung WF, Zhang RQ, Fung YF, Poon RWS, Lam JY, Tam S, Hung IFN, Chen H, Yuen KY, To KKW. Low population serum microneutralization antibody titer against the predominating influenza A(H3N2) N121K virus during the severe influenza summer peak of Hong Kong in 2017. Emerg Microbes Infect 2018; 7:23. [PMID: 29511175 PMCID: PMC5841213 DOI: 10.1038/s41426-018-0041-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 12/13/2022]
Abstract
The 2017 Hong Kong influenza A(H3N2) summer season was unexpectedly severe. However, antigenic characterization of the 2017 circulating A(H3N2) viruses using ferret antisera did not show significant antigenic drift. We analyzed the hemagglutinin amino acid sequences of A(H3N2) virus circulating in Hong Kong in 2017, and found that viruses with hemagglutinin N121K substitution, which was rare before 2017, emerged rapidly and dominated in 2017 (52.4% of A[H3N2] virus in 2017 contains N121K substitution). Microneutralization assay using archived human sera collected from mid-2017 showed that the geometric mean microneutralization titer was 3.6-fold lower against a 2017 cell culture-grown circulating A(H3N2)-N121K virus (3391/2017 virus) than that against the cell culture-grown 2016-2017 A(H3N2) seasonal influenza vaccine-like vaccine virus (4801/2014 virus) (13.4 vs 41.8, P < 0.0001). Significantly fewer serum specimens had a microneutralization titer of 40 or above against 3391/2017 virus than that against 4801/2014 virus (26.4% vs 60.0%, P < 0.0001). Conversely, the geometric mean hemagglutination inhibition titer was slightly higher against 3391/2017 virus than that against the 4801/2014 virus (96.9 vs 55.4, P < 0.0001). Moreover, 59.1% of specimens had a significantly lower microneutralization antibody titer (≥4-fold) against 3391/2017 virus than that against 4801/2014 virus, but none for hemagglutination titer (P < 0.0001). Similar results of microneutralization and hemagglutination titers were observed for day 21-post-vaccination sera. Hence, the 2017 A(H3N2) summer peak in Hong Kong was associated with a low-microneutralization titer against the circulating virus. Our results support the use of microneutralization assay with human serum in assessing population susceptibility and antigenic changes of A(H3N2) virus. Novel and available immunization approach, such as topical imiquimod followed by intradermal vaccination, to broaden the neutralizing antibody response of influenza vaccine should be considered.
Collapse
Affiliation(s)
- Houshun Zhu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Andrew C Y Lee
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Can Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Winger W N Mak
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yetta Y Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
- State Key Laboratory for Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Anna J X Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wai-Fong Fung
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Rui-Qi Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yim-Fong Fung
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Rosana W S Poon
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Joy-Yan Lam
- Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Sidney Tam
- Division of Clinical Biochemistry, Department of Pathology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Ivan F N Hung
- State Key Laboratory for Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Honglin Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- State Key Laboratory for Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China.
- State Key Laboratory for Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
- Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Kelvin K W To
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China.
- State Key Laboratory for Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
- Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
28
|
Gallinaro A, Borghi M, Bona R, Grasso F, Calzoletti L, Palladino L, Cecchetti S, Vescio MF, Macchia D, Morante V, Canitano A, Temperton N, Castrucci MR, Salvatore M, Michelini Z, Cara A, Negri D. Integrase Defective Lentiviral Vector as a Vaccine Platform for Delivering Influenza Antigens. Front Immunol 2018; 9:171. [PMID: 29459873 PMCID: PMC5807328 DOI: 10.3389/fimmu.2018.00171] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 01/19/2018] [Indexed: 12/31/2022] Open
Abstract
Viral vectors represent an attractive technology for vaccine delivery. We exploited the integrase defective lentiviral vector (IDLV) as a platform for delivering relevant antigens within the context of the ADITEC collaborative research program. In particular, Influenza virus hemagglutinin (HA) and nucleoprotein (NP) were delivered by IDLVs while H1N1 A/California/7/2009 subunit vaccine (HAp) with or without adjuvant was used to compare the immune response in a murine model of immunization. In order to maximize the antibody response against HA, both IDLVs were also pseudotyped with HA (IDLV-HA/HA and IDLV-NP/HA, respectively). Groups of CB6F1 mice were immunized intramuscularly with a single dose of IDLV-NP/HA, IDLV-HA/HA, HAp alone, or with HAp together with the systemic adjuvant MF59. Six months after the vaccine prime all groups were boosted with HAp alone. Cellular and antibody responses to influenza antigens were measured at different time points after the immunizations. Mice immunized with HA-pseudotyped IDLVs showed similar levels of anti-H1N1 IgG over time, evaluated by ELISA, which were comparable to those induced by HAp + MF59 vaccination, but significantly higher than those induced by HAp alone. The boost with HAp alone induced an increase of antibodies in all groups, and the responses were maintained at higher levels up to 18 weeks post-boost. The antibody response was functional and persistent overtime, capable of neutralizing virus infectivity, as evaluated by hemagglutination inhibition and microneutralization assays. Moreover, since neuraminidase (NA)-expressing plasmid was included during IDLV preparation, immunization with IDLV-NP/HA and IDLV-HA/HA also induced functional anti-NA antibodies, evaluated by enzyme-linked lectin assay. IFNγ-ELISPOT showed evidence of HA-specific response in IDLV-HA/HA immunized animals and persistent NP-specific CD8+ T cell response in IDLV-NP/HA immunized mice. Taken together our results indicate that IDLV can be harnessed for producing a vaccine able to induce a comprehensive immune response, including functional antibodies directed toward HA and NA proteins present on the vector particles in addition to a functional T cell response directed to the protein transcribed from the vector.
Collapse
Affiliation(s)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta Bona
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Felicia Grasso
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Calzoletti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Serena Cecchetti
- Confocal Microscopy Unit NMR, Confocal Microscopy Area Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Daniele Macchia
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Valeria Morante
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Kent, United Kingdom
| | | | - Mirella Salvatore
- Department of Medicine, Weill Cornell Medical College, New York, United States
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
29
|
Ward BJ, Pillet S, Charland N, Trepanier S, Couillard J, Landry N. The establishment of surrogates and correlates of protection: Useful tools for the licensure of effective influenza vaccines? Hum Vaccin Immunother 2018; 14:647-656. [PMID: 29252098 PMCID: PMC5861778 DOI: 10.1080/21645515.2017.1413518] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The search for a test that can predict vaccine efficacy is an important part of any vaccine development program. Although regulators hesitate to acknowledge any test as a true ‘correlate of protection’, there are many precedents for defining ‘surrogate’ assays. Surrogates can be powerful tools for vaccine optimization, licensure, comparisons between products and development of improved products. When such tests achieve ‘reference’ status however, they can inadvertently become barriers to new technologies that do not work the same way as existing vaccines. This is particularly true when these tests are based upon circularly-defined ‘reference’ or, even worse, proprietary reagents. The situation with inactivated influenza vaccines is a good example of this phenomenon. The most frequently used tests to define vaccine-induced immunity are all serologic assays: hemagglutination inhibition (HI), single radial hemolysis (SRH) and microneutralization (MN). The first two, and particularly the HI assay, have achieved reference status and criteria have been established in many jurisdictions for their use in licensing new vaccines and to compare the performance of different vaccines. However, all of these assays are based on biological reagents that are notoriously difficult to standardize and can vary substantially by geography, by chance (i.e. developing reagents in eggs that may not antigenitically match wild-type viruses) and by intention (ie: choosing reagents that yield the most favorable results). This review describes attempts to standardize these assays to improve their performance as surrogates, the dangers of over-reliance on ‘reference’ serologic assays, the ways that manufacturers can exploit the existing regulatory framework to make their products ‘look good’ and the implications of this long-established system for the introduction of novel influenza vaccines.
Collapse
Affiliation(s)
- Brian J Ward
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | - Stephane Pillet
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | | | | | | | | |
Collapse
|
30
|
TAVAKOLI A, REZAEI F, FATEMI NASAB GS, ADJAMINEZHAD-FARD F, NOROOZBABAEI Z, MOKHTARI-AZAD T. The Comparison of Sensitivity and Specificity of ELISA-based Microneutralization Test with Hemagglutination Inhibition Test to Evaluate Neutralizing Antibody against Influenza Virus (H1N1). IRANIAN JOURNAL OF PUBLIC HEALTH 2017; 46:1690-1696. [PMID: 29259944 PMCID: PMC5734969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The most common serological assay to measure anti-influenza antibodies is hemagglutination inhibition (HI) assay. Recently, neutralizing antibodies against influenza virus infection or vaccination can also be detected using microneutralization assays and occasionally, have greater sensitivity than the standard HI assays. The study aimed to compare the sensitivity and specificity of ELISA-based microneutralization (microNT-ELISA) and conventional HI assays in order to detect influenza H1N1 virus antibodies. METHODS MicroNT-ELISA was set up according to the WHO Manual on Influenza Diagnosis and Surveillance in Virology Department of Tehran University of Medical Sciences for the detection of neutralizing antibodies against H1N1 influenza virus in 2013. Fifty serum samples were analyzed with both HI and microNT-ELISA assays. Correlation between methods was calculated by linear regression analysis. RESULTS The linear correlation coefficient squares, R2, of microNT-ELISA and HI test was 0.61 (P<0.0001) and we observed a high index of coincidence between the two tests. According to McNemar's test, there was no statistically significant difference between these two assays (P>0.05). CONCLUSION The sensitivity and specificity of microNT-ELISA assay were high (87% and 73%, respectively) and closely related to gold standard test results. Therefore, microNT-ELISA is recommended as an alternative or complementary test to conventional HI assay for serological and epidemiological purposes.
Collapse
|
31
|
Gross FL, Bai Y, Jefferson S, Holiday C, Levine MZ. Measuring Influenza Neutralizing Antibody Responses to A(H3N2) Viruses in Human Sera by Microneutralization Assays Using MDCK-SIAT1 Cells. J Vis Exp 2017:56448. [PMID: 29286446 PMCID: PMC5755475 DOI: 10.3791/56448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Neutralizing antibodies against hemagglutinin (HA) of influenza viruses are considered the main immune mechanism that correlates with protection for influenza infections. Microneutralization (MN) assays are often used to measure neutralizing antibody responses in human sera after influenza vaccination or infection. Madine Darby Canine Kidney (MDCK) cells are the commonly used cell substrate for MN assays. However, currently circulating 3C.2a and 3C.3a A(H3N2) influenza viruses have acquired altered receptor binding specificity. The MDCK-SIAT1 cell line with increased α-2,6 sialic galactose moieties on the surface has proven to provide improved infectivity and more faithful replications than conventional MDCK cells for these contemporary A(H3N2) viruses. Here, we describe a MN assay using MDCK-SIAT1 cells that has been optimized to quantify neutralizing antibody titers to these contemporary A(H3N2) viruses. In this protocol, heat inactivated sera containing neutralizing antibodies are first serially diluted, then incubated with 100 TCID50/well of influenza A(H3N2) viruses to allow antibodies in the sera to bind to the viruses. MDCK-SIAT1 cells are then added to the virus-antibody mixture, and incubated for 18 - 20 h at 37 °C, 5% CO2 to allow A(H3N2) viruses to infect MDCK-SIAT1 cells. After overnight incubation, plates are fixed and the amount of virus in each well is quantified by an enzyme-linked immunosorbent assay (ELISA) using anti-influenza A nucleoprotein (NP) monoclonal antibodies. Neutralizing antibody titer is defined as the reciprocal of the highest serum dilution that provides ≥50% inhibition of virus infectivity.
Collapse
Affiliation(s)
- F Liaini Gross
- Influenza Division, Centers for Disease Control and Prevention; Battelle
| | - Yaohui Bai
- Influenza Division, Centers for Disease Control and Prevention
| | | | - Crystal Holiday
- Influenza Division, Centers for Disease Control and Prevention
| | - Min Z Levine
- Influenza Division, Centers for Disease Control and Prevention;
| |
Collapse
|
32
|
Smith G, Liu Y, Flyer D, Massare MJ, Zhou B, Patel N, Ellingsworth L, Lewis M, Cummings JF, Glenn G. Novel hemagglutinin nanoparticle influenza vaccine with Matrix-M™ adjuvant induces hemagglutination inhibition, neutralizing, and protective responses in ferrets against homologous and drifted A(H3N2) subtypes. Vaccine 2017; 35:5366-5372. [PMID: 28844407 DOI: 10.1016/j.vaccine.2017.08.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/05/2017] [Accepted: 08/07/2017] [Indexed: 11/19/2022]
Abstract
Influenza viruses frequently acquire mutations undergoing antigenic drift necessitating annual evaluation of vaccine strains. Highly conserved epitopes have been identified in the hemagglutinin (HA) head and stem regions, however, current influenza vaccines induce only limited responses to these conserved sites. Here, we describe a novel seasonal recombinant HA nanoparticle influenza vaccine (NIV) formulated with a saponin-based adjuvant, Matrix-M™. NIV induced hemagglutination inhibition (HAI) and microneutralizing (MN) antibodies against a broad range of influenza A(H3N2) subtypes. In a comparison of NIV against standard-dose and high-dose inactivated influenza vaccines (IIV and IIV-HD, respectively) in ferrets NIV elicited HAI and MN responses exceeding those induced by IIV-HD against homologous A(H3N2) by 7 fold, A(H1N1) by 26 fold, and B strain viruses by 2 fold. NIV also induced MN responses against all historic A/H3N2 strains tested, spanning more than a decade of viral evolution from the 2000-2017 influenza seasons whereas IIV and IIV-HD induced HAI and MN responses were largely directed against the homologous A(H3N2), A(H1N1), and B virus strains. NIV induced superior protection compared to IIV and IIV-HD in ferrets challenged with a homologous or 10-year drifted influenza A(H3N2) strain. HAI positive and HAI negative neutralizing monoclonal antibodies derived from mice immunized with NIV were active against homologous and drifted influenza A(H3N2) strains. Taken together these observations suggest that NIV can induce responses to one or more highly conserved HA head and stem epitopes and result in highly neutralizing antibodies against both homologous and drift strains.
Collapse
Affiliation(s)
- Gale Smith
- Novavax, Inc., 20 Firstfield Road, Gaithersburg, MD 20878, USA.
| | - Ye Liu
- Novavax, Inc., 20 Firstfield Road, Gaithersburg, MD 20878, USA.
| | - David Flyer
- Novavax, Inc., 20 Firstfield Road, Gaithersburg, MD 20878, USA.
| | | | - Bin Zhou
- Novavax, Inc., 20 Firstfield Road, Gaithersburg, MD 20878, USA.
| | - Nita Patel
- Novavax, Inc., 20 Firstfield Road, Gaithersburg, MD 20878, USA.
| | | | - Maggie Lewis
- Novavax, Inc., 20 Firstfield Road, Gaithersburg, MD 20878, USA.
| | | | - Greg Glenn
- Novavax, Inc., 20 Firstfield Road, Gaithersburg, MD 20878, USA.
| |
Collapse
|
33
|
Truelove S, Zhu H, Lessler J, Riley S, Read JM, Wang S, Kwok KO, Guan Y, Jiang CQ, Cummings DAT. A comparison of hemagglutination inhibition and neutralization assays for characterizing immunity to seasonal influenza A. Influenza Other Respir Viruses 2016; 10:518-524. [PMID: 27406695 PMCID: PMC5059953 DOI: 10.1111/irv.12408] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Serum antibody to influenza can be used to identify past exposure and measure current immune status. The two most common methods for measuring this are the hemagglutination inhibition assay (HI) and the viral neutralization assay (NT), which have not been systematically compared for a large number of influenza viruses. METHODS A total of 151 study participants from near Guangzhou, China, were enrolled in 2009 and provided serum. HI and NT assays were performed for 12 historic and recently circulating strains of seasonal influenza A. We compared titers using Spearman correlation and fit models to predict NT using HI results. RESULTS We observed high positive mean correlation between HI and NT assays (Spearman's rank correlation, ρ=.86) across all strains. Correlation was highest within subtypes and within close proximity in time. Overall, an HI=20 corresponded to NT=10, and HI=40 corresponded to NT=20. Linear regression of log(NT) on log(HI) was statistically significant, with age modifying this relationship. Strain-specific area under a curve (AUC) indicated good accuracy (>80%) for predicting NT with HI. CONCLUSIONS While we found high overall correspondence of titers between NT and HI assays for seasonal influenza A, no exact equivalence between assays could be determined. This was further complicated by correspondence between titers changing with age. These findings support generalized comparison of results between assays and give further support for use of the hemagglutination inhibition assay over the more resource intensive viral neutralization assay for seasonal influenza A, although attention should be given to the effect of age on these assays.
Collapse
Affiliation(s)
- Shaun Truelove
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, 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 SAR, China
| | - Justin Lessler
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Steven Riley
- School of Public Health, Imperial College, London, UK
| | - Jonathan M Read
- Department of Epidemiology and Public Health, Institute of Infection and Global Health, University of Liverpool, Neston, UK
| | - Shuying Wang
- Guangzhou No. 12 Hospital, Guangzhou, Guangdong, China
| | - Kin On Kwok
- 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 SAR, China
| | - Yi Guan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, 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 SAR, China
| | | | - Derek A T Cummings
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Biology, University of Florida, Gainesville, FL, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
34
|
Standardization of Hemagglutination Inhibition Assay for Influenza Serology Allows for High Reproducibility between Laboratories. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:236-42. [PMID: 26818953 DOI: 10.1128/cvi.00613-15] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/14/2016] [Indexed: 11/20/2022]
Abstract
Standardization of the hemagglutination inhibition (HAI) assay for influenza serology is challenging. Poor reproducibility of HAI results from one laboratory to another is widely cited, limiting comparisons between candidate vaccines in different clinical trials and posing challenges for licensing authorities. In this study, we standardized HAI assay materials, methods, and interpretive criteria across five geographically dispersed laboratories of a multidisciplinary influenza research network and then evaluated intralaboratory and interlaboratory variations in HAI titers by repeatedly testing standardized panels of human serum samples. Duplicate precision and reproducibility from comparisons between assays within laboratories were 99.8% (99.2% to 100%) and 98.0% (93.3% to 100%), respectively. The results for 98.9% (95% to 100%) of the samples were within 2-fold of all-laboratory consensus titers, and the results for 94.3% (85% to 100%) of the samples were within 2-fold of our reference laboratory data. Low-titer samples showed the greatest variability in comparisons between assays and between sites. Classification of seroprotection (titer ≥ 40) was accurate in 93.6% or 89.5% of cases in comparison to the consensus or reference laboratory classification, respectively. This study showed that with carefully chosen standardization processes, high reproducibility of HAI results between laboratories is indeed achievable.
Collapse
|
35
|
Eichelberger MC, Couzens L, Gao Y, Levine M, Katz J, Wagner R, Thompson CI, Höschler K, Laurie K, Bai T, Engelhardt OG, Wood J. Comparability of neuraminidase inhibition antibody titers measured by enzyme-linked lectin assay (ELLA) for the analysis of influenza vaccine immunogenicity. Vaccine 2015; 34:458-465. [PMID: 26707221 DOI: 10.1016/j.vaccine.2015.12.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/04/2015] [Accepted: 12/05/2015] [Indexed: 12/31/2022]
Abstract
Neuraminidase-inhibition (NI) antibody titers can be used to evaluate the immunogenicity of inactivated influenza vaccines and have provided evidence of serologic cross-reactivity between seasonal and pandemic H1N1 viruses. The traditional thiobarbituric acid assay is impractical for large serologic analyses, and therefore many laboratories use an enzyme-linked lectin assay (ELLA) to determine serum NI antibody titers. The comparability of ELLA NI antibody titers when measured in different laboratories was unknown. Here we report a study conducted through the Consortium for the Standardisation of Influenza SeroEpidemiology (CONSISE) to evaluate the variability of the ELLA. NI antibody titers of a set of 12 samples were measured against both N1 and N2 neuraminidase antigens in 3 independent assays by each of 23 laboratories. For a sample repeated in the same assay, ≥96% of N1 and N2 assays had less than a 4-fold difference in titer. Comparison of the titers measured in assays conducted on 3 different days in the same laboratory showed that a four-fold difference in titer was uncommon. Titers of the same sera measured in different laboratories spanned 3 to 6 two-fold dilutions (i.e., 8-64 fold difference in titer), with an average percent geometric coefficient of variation (%GCV) of 112 and 82% against N1 and N2 antigens, respectively. The difference in titer as indicated by fold range and %GCV was improved by normalizing the NI titers to a standard that was included in each assay. This study identified background signal and the amount of antigen in the assay as critical factors that influence titer, providing important information toward development of a consensus ELLA protocol.
Collapse
Affiliation(s)
- Maryna C Eichelberger
- Division of Viral Products, CBER, Food and Drug Administration, Silver Spring, MD, USA.
| | - Laura Couzens
- Division of Viral Products, CBER, Food and Drug Administration, Silver Spring, MD, USA
| | - Yonghong Gao
- BARDA, Department of Health and Human Services, Washington, DC, USA
| | - Min Levine
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jacqueline Katz
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | | | | | - Tian Bai
- WHO Collaborating Center, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Othmar G Engelhardt
- National Institute for Biological Standards and Control, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | | | - John Wood
- Formerly National Institute for Biological Standards and Control, United Kingdom
| |
Collapse
|