1
|
Garjani A, Chegini AM, Salehi M, Tabibzadeh A, Yousefi P, Razizadeh MH, Esghaei M, Esghaei M, Rohban MH. Forecasting influenza hemagglutinin mutations through the lens of anomaly detection. Sci Rep 2023; 13:14944. [PMID: 37696867 PMCID: PMC10495359 DOI: 10.1038/s41598-023-42089-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/05/2023] [Indexed: 09/13/2023] Open
Abstract
The influenza virus hemagglutinin is an important part of the virus attachment to the host cells. The hemagglutinin proteins are one of the genetic regions of the virus with a high potential for mutations. Due to the importance of predicting mutations in producing effective and low-cost vaccines, solutions that attempt to approach this problem have recently gained significant attention. A historical record of mutations has been used to train predictive models in such solutions. However, the imbalance between mutations and preserved proteins is a big challenge for the development of such models that need to be addressed. Here, we propose to tackle this challenge through anomaly detection (AD). AD is a well-established field in Machine Learning (ML) that tries to distinguish unseen anomalies from normal patterns using only normal training samples. By considering mutations as anomalous behavior, we could benefit existing rich solutions in this field that have emerged recently. Such methods also fit the problem setup of extreme imbalance between the number of unmutated vs. mutated training samples. Motivated by this formulation, our method tries to find a compact representation for unmutated samples while forcing anomalies to be separated from the normal ones. This helps the model to learn a shared unique representation between normal training samples as much as possible, which improves the discernibility and detectability of mutated samples from the unmutated ones at the test time. We conduct a large number of experiments on four publicly available datasets, consisting of three different hemagglutinin protein datasets, and one SARS-CoV-2 dataset, and show the effectiveness of our method through different standard criteria.
Collapse
Affiliation(s)
- Ali Garjani
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Mohammadreza Salehi
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran
| | - Alireza Tabibzadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parastoo Yousefi
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Moein Esghaei
- Cognitive Neuroscience Laboratory, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
| | - Maryam Esghaei
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
2
|
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
|
3
|
Deng X, Chen Z, Zhao Z, Chen J, Li M, Yang J, Yu H. Regional characteristics of influenza seasonality patterns in mainland China, 2005-2017: a statistical modeling study. Int J Infect Dis 2023; 128:91-97. [PMID: 36581188 DOI: 10.1016/j.ijid.2022.12.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/06/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES To quantify the seasonal and antigenic characteristics of influenza to help understand influenza activity and inform vaccine recommendations. METHODS We employed a generalized linear model with harmonic terms to quantify the seasonal pattern of influenza in China from 2005-2017, including amplitude (circulatory intensity), semiannual periodicity (given two peaks a year), annual peak time, and epidemic duration. The antigenic differences were distinguished as antigenic similarity between 2009 and 2020. We categorized regions above 33° N, between 27° N and 33° N, and below 27° N as the north, central, and south regions, respectively. RESULTS We estimated that the amplitude in the north region (median: 0.019, 95% CI: 0.018-0.021) was significantly higher than that in the central region (median: 0.011, 95% CI: 0.01-0.012, P <0.001) and south region (median: 0.008, 95% CI: 0.007-0.008, P <0.001) for influenza A virus subtype H3N2 (A/H3N2). The A/H3N2 in the central region had a semiannual periodicity (median: 0.548, 95% CI: 0.517-0.577), while no semiannual pattern was found in other regions or subtypes/lineages. The antigenic similarity was low (below 50% in the 2009-2010, 2014-2015, 2016-2018, and 2019-2020 seasons) for A/H3N2. CONCLUSION Our study depicted the seasonal pattern differences and antigenic differences of influenza in China, which provides information for vaccination strategies.
Collapse
Affiliation(s)
- Xiaowei Deng
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Zhiyuan Chen
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Zeyao Zhao
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Junbo Chen
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Mei Li
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Juan Yang
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Hongjie Yu
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China; National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Patel MC, Flanigan D, Feng C, Chesnokov A, Nguyen HT, Elal AA, Steel J, Kondor RJ, Wentworth DE, Gubareva LV, Mishin VP. An optimized cell-based assay to assess influenza virus replication by measuring neuraminidase activity and its applications for virological surveillance. Antiviral Res 2022; 208:105457. [PMID: 36332755 PMCID: PMC10149149 DOI: 10.1016/j.antiviral.2022.105457] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/12/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
Year-round virological characterization of circulating epidemic influenza viruses is conducted worldwide to detect the emergence of viruses that may escape pre-existing immunity or acquire resistance to antivirals. High throughput phenotypic assays are needed to complement the sequence-based analysis of circulating viruses and improve pandemic preparedness. The recent entry of a polymerase inhibitor, baloxavir, into the global market further highlighted this need. Here, we optimized a cell-based assay that considerably streamlines antiviral and antigenic testing by replacing lengthy immunostaining and imaging procedures used in current assay with measuring the enzymatic activity of nascent neuraminidase (NA) molecules expressed on the surface of virus-infected cells. For convenience, this new assay was named IRINA (Influenza Replication Inhibition Neuraminidase-based Assay). IRINA was successfully validated to assess inhibitory activity of baloxavir on virus replication by testing a large set (>150) of influenza A and B viruses, including drug resistant strains and viruses collected during 2017-2022. To test its versatility, IRINA was utilized to evaluate neutralization activity of a broadly reactive human anti-HA monoclonal antibody, FI6, and post-infection ferret antisera, as well as the inhibition of NA enzyme activity by NA inhibitors. Performance of IRINA was tested in parallel using respective conventional assays. IRINA offers an attractive alternative to current phenotypic assays, while maintaining reproducibility and high throughput capacity. Additionally, the improved turnaround time may prove to be advantageous when conducting time sensitive studies, such as investigating a new virus outbreak. This assay can meet the needs of surveillance laboratories by providing a streamlined and cost-effective approach for virus characterization.
Collapse
Affiliation(s)
- Mira C Patel
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Daniel Flanigan
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; General Dynamics Information Technology, Atlanta, GA, USA
| | - Chenchen Feng
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Anton Chesnokov
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ha T Nguyen
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Anwar Abd Elal
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA; Cherokee Nation Integrated Health, L.L.C., Atlanta, GA, USA
| | - John Steel
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Rebecca J Kondor
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - David E Wentworth
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Larisa V Gubareva
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Vasiliy P Mishin
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
5
|
Frische A, Brooks PT, Gybel-Brask M, Sækmose SG, Jensen BA, Mikkelsen S, Bruun MT, Boding L, Strandh CP, Jørgensen CS, Krogfelt KA, Fomsgaard A, Lassauniere R. Optimization and evaluation of a live virus SARS-CoV-2 neutralization assay. PLoS One 2022; 17:e0272298. [PMID: 35901110 PMCID: PMC9333216 DOI: 10.1371/journal.pone.0272298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/15/2022] [Indexed: 12/23/2022] Open
Abstract
Virus neutralization assays provide a means to quantitate functional antibody responses that block virus infection. These assays are instrumental in defining vaccine and therapeutic antibody potency, immune evasion by viral variants, and post-infection immunity. Here we describe the development, optimization and evaluation of a live virus microneutralization assay specific for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this assay, SARS-CoV-2 clinical isolates are pre-incubated with serial diluted antibody and added to Vero E6 cells. Replicating virus is quantitated by enzyme-linked immunosorbent assay (ELISA) targeting the SARS-CoV-2 nucleocapsid protein and the standardized 50% virus inhibition titer calculated. We evaluated critical test parameters that include virus titration, assay linearity, number of cells, viral dose, incubation period post-inoculation, and normalization methods. Virus titration at 96 hours was determined optimal to account for different growth kinetics of clinical isolates. Nucleocapsid protein levels directly correlated with virus inoculum, with the strongest correlation at 24 hours post-inoculation. Variance was minimized by infecting a cell monolayer, rather than a cell suspension. Neutralization titers modestly decreased with increasing numbers of Vero E6 cells and virus amount. Application of two different normalization models effectively reduced the intermediate precision coefficient of variance to <16.5%. The SARS-CoV-2 microneutralization assay described and evaluated here is based on the influenza virus microneutralization assay described by WHO, and are proposed as a standard assay for comparing neutralization investigations.
Collapse
Affiliation(s)
- Anders Frische
- Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | | | - Mikkel Gybel-Brask
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Susan Mikkelsen
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Mie Topholm Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Lasse Boding
- Danish National Biobank, Statens Serum Institut, Copenhagen, Denmark
| | - Charlotta Polacek Strandh
- Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | | | - Karen Angeliki Krogfelt
- Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
- Department of Molecular and Medicinal Biology, Institute for Science and Environment, Roskilde University, Roskilde, Denmark
| | - Anders Fomsgaard
- Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
- Infectious Diseases Unit, Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - Ria Lassauniere
- Department of Virus & Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
6
|
Ge J, Lin X, Guo J, Liu L, Li Z, Lan Y, Liu L, Guo J, Lu J, Huang W, Xin L, Wang D, Qin K, Xu C, Zhou J. The Antibody Response Against Neuraminidase in Human Influenza A (H3N2) Virus Infections During 2018/2019 Flu Season: Focusing on the Epitopes of 329- N-Glycosylation and E344 in N2. Front Microbiol 2022; 13:845088. [PMID: 35387078 PMCID: PMC8978628 DOI: 10.3389/fmicb.2022.845088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/10/2022] [Indexed: 11/13/2022] Open
Abstract
Seasonal influenza A (H3N2) virus has been a concern since its first introduction in humans in 1968. Accumulating antigenic changes in viral hemagglutinin (HA), particularly recent cocirculations of multiple HA genetic clades, allow H3N2 virus evade into humans annually. From 2010, the binding of neuraminidase (NA) to sialic acid made the traditional assay for HA inhibition antibodies (Abs) unsuitable for antigenicity characterization. Here, we investigated the serum anti-NA response in a cohort with a seroconversion of microneutralizing (MN) Abs targeting the circulating strain, A/Singapore/INFIMH-16-0019/2016 (H3N2, 3C.2a1)-like, a virus during 2018/2019 flu seasons. We discovered that MN Ab titers show no difference between children and adults. Nevertheless, higher titers of Abs with NA activity inhibition (NI) activity of 129 and seroconversion rate of 68.42% are presented in children aged 7-17 years (n = 19) and 73.47 and 41.17% in adults aged 21-59 years (n = 17), respectively. The MN Abs generated in children display direct correlations with HA- and NA-binding Abs or NI Abs. The NI activity exhibited cross-reactivity to N2 of H3N2 viruses of 2007 and 2013, commonly with 329-N-glycosylation and E344 in N2, a characteristic of earlier 3C.2a H3N2 virus in 2014. The percentage of such viruses pronouncedly decreased and was even replaced by those dominant H3N2 viruses with E344K and 329 non-glycosylation, which have a significantly low activity to the tested antisera. Our findings suggest that NI assay is a testable assay applied in H3N2 infection in children, and the antigenic drift of current N2 should be considered for vaccine selection.
Collapse
Affiliation(s)
- Jing Ge
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Xiaojing Lin
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Jinlei Guo
- The Disease Control and Prevention of Qinhuai District, Nanjing, China
| | - Ling Liu
- Qinhuai District Center for Disease Control and Prevention, Nanjing, China
| | - Zi Li
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Yu Lan
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Liqi Liu
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Junfeng Guo
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Jian Lu
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Weijuan Huang
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Li Xin
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Dayan Wang
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Kun Qin
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Cuiling Xu
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Jianfang Zhou
- Key Laboratory for Medical Virology, National Health, and Family Planning Commission, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, Beijing, China
| |
Collapse
|
7
|
Rosadas C, Khan M, Parker E, Marchesin F, Katsanovskaja K, Sureda-Vives M, Fernandez N, Randell P, Harvey R, Lilley A, Harris BHL, Zuhair M, Fertleman M, Ijaz S, Dicks S, Short CE, Quinlan R, Taylor GP, Hu K, McKay P, Rosa A, Roustan C, Zuckerman M, El Bouzidi K, Cooke G, Flower B, Moshe M, Elliott P, Spencer AJ, Lambe T, Gilbert SC, Kingston H, Baillie JK, Openshaw PJM, Semple MG, Cherepanov P, McClure MO, Tedder RS. Detection and quantification of antibody to SARS CoV 2 receptor binding domain provides enhanced sensitivity, specificity and utility. J Virol Methods 2022; 302:114475. [PMID: 35077719 PMCID: PMC8782753 DOI: 10.1016/j.jviromet.2022.114475] [Citation(s) in RCA: 2] [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] [Received: 11/03/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/10/2023]
Abstract
Accurate and sensitive detection of antibody to SARS-CoV-2 remains an essential component of the pandemic response. Measuring antibody that predicts neutralising activity and the vaccine response is an absolute requirement for laboratory-based confirmatory and reference activity. The viral receptor binding domain (RBD) constitutes the prime target antigen for neutralising antibody. A double antigen binding assay (DABA), providing the most sensitive format has been exploited in a novel hybrid manner employing a solid-phase S1 preferentially presenting RBD, coupled with a labelled RBD conjugate, used in a two-step sequential assay for detection and measurement of antibody to RBD (anti-RBD). This class and species neutral assay showed a specificity of 100 % on 825 pre COVID-19 samples and a potential sensitivity of 99.6 % on 276 recovery samples, predicting quantitatively the presence of neutralising antibody determined by pseudo-type neutralization and by plaque reduction. Anti-RBD is also measurable in ferrets immunised with ChadOx1 nCoV-19 vaccine and in humans immunised with both AstraZeneca and Pfizer vaccines. This assay detects anti-RBD at presentation with illness, demonstrates its elevation with disease severity, its sequel to asymptomatic infection and its persistence after the loss of antibody to the nucleoprotein (anti-NP). It also provides serological confirmation of prior infection and offers a secure measure for seroprevalence and studies of vaccine immunisation in human and animal populations. The hybrid DABA also displays the attributes necessary for the detection and quantification of anti-RBD to be used in clinical practice. An absence of detectable anti-RBD by this assay predicates the need for passive immune prophylaxis in at-risk patients.
Collapse
Affiliation(s)
- Carolina Rosadas
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Maryam Khan
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Eleanor Parker
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Federica Marchesin
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Ksenia Katsanovskaja
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Macià Sureda-Vives
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Natalia Fernandez
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Paul Randell
- Department of Infection and Immunity, Imperial College Healthcare NHS Trust, Charing Cross Hospital, London, W6 8RF, UK
| | - Ruth Harvey
- Worldwide Influenza Centre, Francis Crick Institute, London, NW1 1AT, UK
| | - Alice Lilley
- Worldwide Influenza Centre, Francis Crick Institute, London, NW1 1AT, UK
| | - Benjamin H L Harris
- The Wellington Hospital, Circus Road, St John's Wood, London, NW8 6PD, UK; Computational Biology and Integrative Genomics, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Mohamed Zuhair
- The Wellington Hospital, Circus Road, St John's Wood, London, NW8 6PD, UK
| | - Michael Fertleman
- The Wellington Hospital, Circus Road, St John's Wood, London, NW8 6PD, UK
| | - Samreen Ijaz
- Blood Borne Virus Unit, National Infection Service, Colindale Public Health England, London, NW9 5EQ, UK
| | - Steve Dicks
- Blood Borne Virus Unit, National Infection Service, Colindale Public Health England, London, NW9 5EQ, UK; Transfusion Microbiology, NHS Blood and Transplant, Lingard Avenue, London, NW9 5BG, UK
| | - Charlotte-Eve Short
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Rachael Quinlan
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Graham P Taylor
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Kai Hu
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Paul McKay
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Annachiara Rosa
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Crick COVID19 Consortium, Francis Crick Institute, London, NW1 1AT, UK
| | - Chloe Roustan
- Structural Biology Science Technology Platform, Francis Crick Institute, London, NW1 1AT, UK; Crick COVID19 Consortium, Francis Crick Institute, London, NW1 1AT, UK
| | - Mark Zuckerman
- Department of Virology, King's College Hospital, London, SE5 9RS, UK
| | - Kate El Bouzidi
- Department of Virology, King's College Hospital, London, SE5 9RS, UK
| | - Graham Cooke
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Barnaby Flower
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Maya Moshe
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | | | - Teresa Lambe
- Jenner Institute, University of Oxford, ORCRB, Oxford, OX3 7DQ, UK
| | - Sarah C Gilbert
- Jenner Institute, University of Oxford, ORCRB, Oxford, OX3 7DQ, UK
| | - Hugh Kingston
- Department of Infection and Immunity, Imperial College Healthcare NHS Trust, Charing Cross Hospital, London, W6 8RF, UK
| | | | - Peter J M Openshaw
- National Heart and Lung Institute, Imperial College London, Chelsea, London, SW3 6LY, UK
| | - Malcolm G Semple
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, L69 7BE, UK
| | - Peter Cherepanov
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK; Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Crick COVID19 Consortium, Francis Crick Institute, London, NW1 1AT, UK
| | - Myra O McClure
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Richard S Tedder
- Department of Infectious Disease, Imperial College London, St Mary's Campus, London, W2 1PG, UK.
| |
Collapse
|
8
|
Wang Y, Tang CY, Wan XF. Antigenic characterization of influenza and SARS-CoV-2 viruses. Anal Bioanal Chem 2022; 414:2841-2881. [PMID: 34905077 PMCID: PMC8669429 DOI: 10.1007/s00216-021-03806-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022]
Abstract
Antigenic characterization of emerging and re-emerging viruses is necessary for the prevention of and response to outbreaks, evaluation of infection mechanisms, understanding of virus evolution, and selection of strains for vaccine development. Primary analytic methods, including enzyme-linked immunosorbent/lectin assays, hemagglutination inhibition, neuraminidase inhibition, micro-neutralization assays, and antigenic cartography, have been widely used in the field of influenza research. These techniques have been improved upon over time for increased analytical capacity, and some have been mobilized for the rapid characterization of the SARS-CoV-2 virus as well as its variants, facilitating the development of highly effective vaccines within 1 year of the initially reported outbreak. While great strides have been made for evaluating the antigenic properties of these viruses, multiple challenges prevent efficient vaccine strain selection and accurate assessment. For influenza, these barriers include the requirement for a large virus quantity to perform the assays, more than what can typically be provided by the clinical samples alone, cell- or egg-adapted mutations that can cause antigenic mismatch between the vaccine strain and circulating viruses, and up to a 6-month duration of vaccine development after vaccine strain selection, which allows viruses to continue evolving with potential for antigenic drift and, thus, antigenic mismatch between the vaccine strain and the emerging epidemic strain. SARS-CoV-2 characterization has faced similar challenges with the additional barrier of the need for facilities with high biosafety levels due to its infectious nature. In this study, we review the primary analytic methods used for antigenic characterization of influenza and SARS-CoV-2 and discuss the barriers of these methods and current developments for addressing these challenges.
Collapse
Affiliation(s)
- Yang Wang
- MU Center for Influenza and Emerging Infectious Diseases (CIEID), University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Cynthia Y Tang
- MU Center for Influenza and Emerging Infectious Diseases (CIEID), University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
| | - Xiu-Feng Wan
- MU Center for Influenza and Emerging Infectious Diseases (CIEID), University of Missouri, Columbia, MO, USA.
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA.
- Department of Electrical Engineering & Computer Science, College of Engineering, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
9
|
Kodama T, Ueno K, Kondo T, Morozumi Y, Kato A, Nagai S, Shibuya K, Sasakawa C. Spectrophotometric microplate assay for titration and neutralization of avian nephritis virus based on the virus cytopathicity. J Virol Methods 2021; 299:114303. [PMID: 34606795 DOI: 10.1016/j.jviromet.2021.114303] [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: 07/20/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Plaque assay (PA) is a gold standard for virus titration and neutralization of various cytopathic viruses, including avian nephritis virus (ANV), the etiological agent associated with kidney disorders in chickens. In this study, as an alternative to the labor-intensive PA, we developed a spectrophotometric microplate assay (MA) for ANV titration and neutralization based on the virus cytopathicity to primary chicken kidney (CK) cells. METHODS CK cells were infected with ANV in the presence or absence of chicken serum in a 96-well microplate, and the virus-induced cytolysis was quantified by measurement of neutral red uptake using a spectrophotometer. The absorbance values obtained were subjected to a sigmoidal four-parameter logistic regression analysis for the virus titer determination and serum neutralization assessment. Accuracy and reliability of the serum neutralization MA in comparison to the standard PA was statistically evaluated. RESULTS The ANV-MA was capable of quantifying infectious virus titers based on a virus dose-dependent cytolysis of CK cells, and serum neutralization could be assessed as an inhibition of the virus-induced cytolysis accordingly. Statistical evaluation using a 2 × 2 contingency table and receiver-operating characteristic analyses showed 82 % sensitivity, 99 % specificity and 0.97 area under the curve, supporting an overall diagnostic accuracy of the neutralization MA. CONCLUSION The newly developed MA using simplified experimental procedures in the microplate format and direct spectophotometric data readout is readily applicable to general laboratories for high-throughput screening of serum neutralization of ANV.
Collapse
Affiliation(s)
- Toshiaki Kodama
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan.
| | - Kosei Ueno
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan
| | - Tomomi Kondo
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan
| | - Yuki Morozumi
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan
| | - Atsushi Kato
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan
| | - Shinya Nagai
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan
| | - Kazumoto Shibuya
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan
| | - Chihiro Sasakawa
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo, 198-0024, Japan; Medical Mycology Research Center, Chiba University, 1-8-1 Inohara, Chiba, 260-8673, Japan
| |
Collapse
|
10
|
Deakin CT, Cornish GH, Ng KW, Faulkner N, Bolland W, Hope J, Rosa A, Harvey R, Hussain S, Earl C, Jebson BR, Wilkinson MGLL, Marshall LR, O'Brien K, Rosser EC, Radziszewska A, Peckham H, Patel H, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Spyer MJ, Gamblin SJ, McCauley J, Nastouli E, Levin M, Cherepanov P, Ciurtin C, Wedderburn LR, Kassiotis G. Favorable antibody responses to human coronaviruses in children and adolescents with autoimmune rheumatic diseases. MED 2021; 2:1093-1109.e6. [PMID: 34414384 PMCID: PMC8363467 DOI: 10.1016/j.medj.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/06/2021] [Accepted: 08/06/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Differences in humoral immunity to coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), between children and adults remain unexplained, and the effect of underlying immune dysfunction or suppression is unknown. Here, we sought to examine the antibody immune competence of children and adolescents with prevalent inflammatory rheumatic diseases, juvenile idiopathic arthritis (JIA), juvenile dermatomyositis (JDM), and juvenile systemic lupus erythematosus (JSLE) against the seasonal human coronavirus (HCoV)-OC43 that frequently infects this age group. METHODS Sera were collected from JIA (n = 118), JDM (n = 49), and JSLE (n = 30) patients and from healthy control (n = 54) children and adolescents prior to the coronavirus disease 19 (COVID-19) pandemic. We used sensitive flow-cytometry-based assays to determine titers of antibodies that reacted with the spike and nucleoprotein of HCoV-OC43 and cross-reacted with the spike and nucleoprotein of SARS-CoV-2, and we compared them with respective titers in sera from patients with multisystem inflammatory syndrome in children and adolescents (MIS-C). FINDINGS Despite immune dysfunction and immunosuppressive treatment, JIA, JDM, and JSLE patients maintained comparable or stronger humoral responses than healthier peers, which was dominated by immunoglobulin G (IgG) antibodies to HCoV-OC43 spike, and harbored IgG antibodies that cross-reacted with SARS-CoV-2 spike. In contrast, responses to HCoV-OC43 and SARS-CoV-2 nucleoproteins exhibited delayed age-dependent class-switching and were not elevated in JIA, JDM, and JSLE patients, which argues against increased exposure. CONCLUSIONS Consequently, autoimmune rheumatic diseases and their treatment were associated with a favorable ratio of spike to nucleoprotein antibodies. FUNDING This work was supported by a Centre of Excellence Centre for Adolescent Rheumatology Versus Arthritis grant, 21593, UKRI funding reference MR/R013926/1, the Great Ormond Street Children's Charity, Cure JM Foundation, Myositis UK, Lupus UK, and the NIHR Biomedical Research Centres at GOSH and UCLH. This work was supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK, the UK Medical Research Council, and the Wellcome Trust.
Collapse
Affiliation(s)
- Claire T Deakin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at GOSH, London, UK
- OPAL Rheumatology Ltd, Sydney, NSW, Australia
| | - Georgina H Cornish
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Kevin W Ng
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nikhil Faulkner
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - William Bolland
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Joshua Hope
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Annachiara Rosa
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Saira Hussain
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Christopher Earl
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Bethany R Jebson
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at GOSH, London, UK
| | - Meredyth G L L Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at GOSH, London, UK
| | - Lucy R Marshall
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at GOSH, London, UK
| | - Kathryn O'Brien
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at GOSH, London, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, UK
| | - Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, UK
| | - Harsita Patel
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | | | | | | | - Catherine F Houlihan
- UCLH NHS Trust, London NW1 2BU, UK
- Division of Infection and Immunity, UCL, London WC1E 6BT, UK
| | - Moira J Spyer
- UCLH NHS Trust, London NW1 2BU, UK
- Department of Population, Policy and Practice, Great Ormond Street ICH, UCL, London WC1N 1EH, UK
| | - Steve J Gamblin
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - John McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Eleni Nastouli
- UCLH NHS Trust, London NW1 2BU, UK
- Department of Population, Policy and Practice, Great Ormond Street ICH, UCL, London WC1N 1EH, UK
| | - Michael Levin
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Infectious Disease, St Mary's Hospital, Imperial College London, London W2 1NY, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, UK
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospitals (UCLH), Great Ormond Street Hospital (GOSH), London, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London, UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at GOSH, London, UK
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Infectious Disease, St Mary's Hospital, Imperial College London, London W2 1NY, UK
| |
Collapse
|
11
|
Lim J, Cheong Y, Kim YS, Chae W, Hwang BJ, Lee J, Jang YH, Roh YH, Seo SU, Seong BL. RNA-dependent assembly of chimeric antigen nanoparticles as an efficient H5N1 pre-pandemic vaccine platform. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102438. [PMID: 34256061 DOI: 10.1016/j.nano.2021.102438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 11/17/2022]
Abstract
Highly pathogenic avian influenza viruses (HPAIVs) pose a significant threat to human health, with high mortality rates, and require effective vaccines. We showed that, harnessed with novel RNA-mediated chaperone function, hemagglutinin (HA) of H5N1 HPAIV could be displayed as an immunologically relevant conformation on self-assembled chimeric nanoparticles (cNP). A tri-partite monomeric antigen was designed including: i) an RNA-interaction domain (RID) as a docking tag for RNA to enable chaperna function (chaperna: chaperone + RNA), ii) globular head domain (gd) of HA as a target antigen, and iii) ferritin as a scaffold for 24 mer-assembly. The immunization of mice with the nanoparticles (~46 nm) induced a 25-30 fold higher neutralizing capacity of the antibody and provided cross-protection from homologous and heterologous lethal challenges. This study suggests that cNP assembly is conducive to eliciting antibodies against the conserved region in HA, providing potent and broad protective efficacy.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Birds/virology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/therapeutic use
- Humans
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/chemistry
- Influenza Vaccines/immunology
- Influenza Vaccines/therapeutic use
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Mice
- Nanoparticles/chemistry
- Nanoparticles/therapeutic use
- Pandemics
- RNA/genetics
- RNA/immunology
- RNA/therapeutic use
Collapse
Affiliation(s)
- Jongkwan Lim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yucheol Cheong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young-Seok Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Wonil Chae
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Beom Jeung Hwang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jinhee Lee
- Department of Integrated OMICS for Biomedical Science, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yo Han Jang
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Biotechnology, Andong National University, Andong, Republic of Korea
| | - Young Hoon Roh
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Baik L Seong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, Yonsei University, Seoul, Republic of Korea; Vaccine Innovative Technology Alliance-Korea, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Wang G, Huang P, Hong J, Fu R, Wu Q, Chen R, Lin L, Han Q, Chen H, Chen Y, Xia N. Establishment of a rapid ELISPOT assay for influenza virus titration and neutralizing antibody detection. J Med Virol 2021; 93:3455-3464. [PMID: 32621615 DOI: 10.1002/jmv.26257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 06/30/2020] [Indexed: 11/05/2022]
Abstract
Seasonal influenza is an acute respiratory infection causing around 500 000 global deaths annually. There is an unmet medical need to develop more effective antiviral drugs and vaccines against influenza infection. A rapid, accurate, high-throughput titration assay for influenza virus particles or neutralizing antibodies would be extremely useful in these research fields. However, commonly used methods such as tissue culture infective dose and plaque-forming units (PFU) for virus particle quantification, and the plaque reduction neutralization test (PRNT) for antibody determination are time-consuming, laborious, and have limited accuracy. In this study, we developed an efficient assay based on the enzyme-linked immunospot (ELISPOT) technique for the influenza virus and neutralizing antibody titration. Two broad-spectrum antibodies recognizing the nucleoproteins of influenza A and B viruses were used in the assay to broadly and highly sensitively detect influenza virus-infected cells at 16 hours postinfection. An optimized cell culture medium with no tosyl phenylalanyl chloromethyl ketone trypsin and high dose oseltamivir acid was used to improve quantitation accuracy. This ELISPOT assay displayed a good correlation (R2 = 0.9851) with the PFU assay when used to titrate 30 influenza virus isolates. The assay was also applied to measure influenza-neutralizing antibodies in 40 human sera samples, showing a good correlation (R2 = 0.9965) with the PRNT assay. This ELISPOT titration assay is a rapid, accurate, high-throughput assay for quantification of influenza virus and neutralizing antibodies, and provides a powerful tool for research into and development of drugs and vaccines against influenza.
Collapse
Affiliation(s)
- Guosong Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Pengfei Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Junping Hong
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Rao Fu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Qian Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Ruiqi Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Lina Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Qiangyuan Han
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Honglin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
| | - Yixin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
13
|
Differential Influence of Age on the Relationship between Genetic Mismatch and A(H1N1)pdm09 Vaccine Effectiveness. Viruses 2021; 13:v13040619. [PMID: 33916601 PMCID: PMC8065480 DOI: 10.3390/v13040619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 01/02/2023] Open
Abstract
Assessment of influenza vaccine effectiveness (VE) and identification of relevant influencing factors are the current priorities for optimizing vaccines to reduce the impacts of influenza. To date, how the difference between epidemic strains and vaccine strains at genetic scale affects age-specific vaccine performance remains ambiguous. This study investigated the association between genetic mismatch on hemagglutinin and neuraminidase genes and A(H1N1)pdm09 VE in different age groups with a novel computational approach. We found significant linear relationships between VE and genetic mismatch in children, young adults, and middle-aged adults. In the children’s group, each 3-key amino acid mutation was associated with an average of 10% decrease in vaccine effectiveness in a given epidemic season, and genetic mismatch exerted no influence on VE for the elderly group. We demonstrated that present vaccines were most effective for children, while protection for the elderly was reduced and indifferent to vaccine component updates. Modeling such relationships is practical to inform timely evaluation of VE in different groups of populations during mass vaccination and may inform age-specific vaccination regimens.
Collapse
|
14
|
Ursin RL, Liu H, Powell HR, Westerbeck JW, Shaw-Saliba K, Sylvia KE, Fenstermacher KJ, Mehoke T, Thielen P, Rothman RE, Pekosz A, Klein SL. Differential Antibody Recognition of H3N2 Vaccine and Seasonal Influenza Virus Strains Based on Age, Vaccine Status, and Sex in the 2017-2018 Season. J Infect Dis 2021; 222:1371-1382. [PMID: 32496543 DOI: 10.1093/infdis/jiaa289] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/22/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND An antigenic mismatch between the vaccine and circulating H3N2 strains was hypothesized to contribute to the severity of the 2017-2018 season in North America. METHODS Serum and nasal washes were collected from influenza positive and negative patients during the 2017-2018 season to determine neutralizing antibody (nAb) titers and for influenza virus sequencing, respectively. RESULTS The circulating and vaccine H3N2 virus strains were different clades, with the vaccine strain being clade 3C.2a and the circulating viruses being 3C.2a2 or 3C.3a. At enrollment, both the H3N2 negative and positive patients had greater nAb titers to the egg-adapted vaccine virus compared to the cell-grown vaccine but the H3N2-negative population had significantly greater titers to the circulating 3C.2a2. Among H3N2-positive patients, vaccination, younger age, and female sex were associated with greater nAb responses to the egg-adapted vaccine H3N2 virus but not to the cell-grown vaccine or circulating viruses. CONCLUSIONS For the 2017-2018 circulating viruses, mutations introduced by egg adaptation decreased vaccine efficacy. No increased protection was afforded by vaccination, younger age, or female sex against 2017-2018 circulating H3N2 viruses.
Collapse
Affiliation(s)
- Rebecca L Ursin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Hsuan Liu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Harrison R Powell
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jason W Westerbeck
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kathryn Shaw-Saliba
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristyn E Sylvia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Katherine J Fenstermacher
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tom Mehoke
- Johns Hopkins Applied Physics Laboratory, Laurel, Maryland, USA
| | - Peter Thielen
- Johns Hopkins Applied Physics Laboratory, Laurel, Maryland, USA
| | - Richard E Rothman
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Huang KYA, Tan TK, Chen TH, Huang CG, Harvey R, Hussain S, Chen CP, Harding A, Gilbert-Jaramillo J, Liu X, Knight M, Schimanski L, Shih SR, Lin YC, Cheng CY, Cheng SH, Huang YC, Lin TY, Jan JT, Ma C, James W, Daniels RS, McCauley JW, Rijal P, Townsend AR. Breadth and function of antibody response to acute SARS-CoV-2 infection in humans. PLoS Pathog 2021; 17:e1009352. [PMID: 33635919 PMCID: PMC8130932 DOI: 10.1371/journal.ppat.1009352] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 05/18/2021] [Accepted: 02/02/2021] [Indexed: 12/31/2022] Open
Abstract
Serological and plasmablast responses and plasmablast-derived IgG monoclonal antibodies (MAbs) have been analysed in three COVID-19 patients with different clinical severities. Potent humoral responses were detected within 3 weeks of onset of illness in all patients and the serological titre was elicited soon after or concomitantly with peripheral plasmablast response. An average of 13.7% and 3.5% of plasmablast-derived MAbs were reactive with virus spike glycoprotein or nucleocapsid, respectively. A subset of anti-spike (10 of 32) antibodies cross-reacted with other betacoronaviruses tested and harboured extensive somatic mutations, indicative of an expansion of memory B cells upon SARS-CoV-2 infection. Fourteen of 32 anti-spike MAbs, including five anti-receptor-binding domain (RBD), three anti-non-RBD S1 and six anti-S2, neutralised wild-type SARS-CoV-2 in independent assays. Anti-RBD MAbs were further grouped into four cross-inhibiting clusters, of which six antibodies from three separate clusters blocked the binding of RBD to ACE2 and five were neutralising. All ACE2-blocking anti-RBD antibodies were isolated from two recovered patients with prolonged fever, which is compatible with substantial ACE2-blocking response in their sera. Finally, the identification of non-competing pairs of neutralising antibodies would offer potential templates for the development of prophylactic and therapeutic agents against SARS-CoV-2.
Collapse
Affiliation(s)
- Kuan-Ying A. Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tiong Kit Tan
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ting-Hua Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Chung-Guei Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, United Kingdom
| | - Saira Hussain
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, United Kingdom
| | - Cheng-Pin Chen
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and National Yang-Ming University, Taipei, Taiwan
| | - Adam Harding
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | - Xu Liu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Michael Knight
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Lisa Schimanski
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Chun Lin
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and Taipei Medical University, Taipei, Taiwan
| | - Chien-Yu Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and National Yang-Ming University, Taipei, Taiwan
| | - Shu-Hsing Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and Taipei Medical University, Taipei, Taiwan
| | - Yhu-Chering Huang
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tzou-Yien Lin
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Che Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - William James
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Rodney S. Daniels
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, United Kingdom
| | - John W. McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, United Kingdom
| | - Pramila Rijal
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
| | - Alain R. Townsend
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
T Cell Immunity against Influenza: The Long Way from Animal Models Towards a Real-Life Universal Flu Vaccine. Viruses 2021; 13:v13020199. [PMID: 33525620 PMCID: PMC7911237 DOI: 10.3390/v13020199] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Current flu vaccines rely on the induction of strain-specific neutralizing antibodies, which leaves the population vulnerable to drifted seasonal or newly emerged pandemic strains. Therefore, universal flu vaccine approaches that induce broad immunity against conserved parts of influenza have top priority in research. Cross-reactive T cell responses, especially tissue-resident memory T cells in the respiratory tract, provide efficient heterologous immunity, and must therefore be a key component of universal flu vaccines. Here, we review recent findings about T cell-based flu immunity, with an emphasis on tissue-resident memory T cells in the respiratory tract of humans and different animal models. Furthermore, we provide an update on preclinical and clinical studies evaluating T cell-evoking flu vaccines, and discuss the implementation of T cell immunity in real-life vaccine policies.
Collapse
|
17
|
Tan TK, Rijal P, Rahikainen R, Keeble AH, Schimanski L, Hussain S, Harvey R, Hayes JWP, Edwards JC, McLean RK, Martini V, Pedrera M, Thakur N, Conceicao C, Dietrich I, Shelton H, Ludi A, Wilsden G, Browning C, Zagrajek AK, Bialy D, Bhat S, Stevenson-Leggett P, Hollinghurst P, Tully M, Moffat K, Chiu C, Waters R, Gray A, Azhar M, Mioulet V, Newman J, Asfor AS, Burman A, Crossley S, Hammond JA, Tchilian E, Charleston B, Bailey D, Tuthill TJ, Graham SP, Duyvesteyn HME, Malinauskas T, Huo J, Tree JA, Buttigieg KR, Owens RJ, Carroll MW, Daniels RS, McCauley JW, Stuart DI, Huang KYA, Howarth M, Townsend AR. A COVID-19 vaccine candidate using SpyCatcher multimerization of the SARS-CoV-2 spike protein receptor-binding domain induces potent neutralising antibody responses. Nat Commun 2021; 12:542. [PMID: 33483491 PMCID: PMC7822889 DOI: 10.1038/s41467-020-20654-7] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/10/2020] [Indexed: 12/18/2022] Open
Abstract
There is need for effective and affordable vaccines against SARS-CoV-2 to tackle the ongoing pandemic. In this study, we describe a protein nanoparticle vaccine against SARS-CoV-2. The vaccine is based on the display of coronavirus spike glycoprotein receptor-binding domain (RBD) on a synthetic virus-like particle (VLP) platform, SpyCatcher003-mi3, using SpyTag/SpyCatcher technology. Low doses of RBD-SpyVLP in a prime-boost regimen induce a strong neutralising antibody response in mice and pigs that is superior to convalescent human sera. We evaluate antibody quality using ACE2 blocking and neutralisation of cell infection by pseudovirus or wild-type SARS-CoV-2. Using competition assays with a monoclonal antibody panel, we show that RBD-SpyVLP induces a polyclonal antibody response that recognises key epitopes on the RBD, reducing the likelihood of selecting neutralisation-escape mutants. Moreover, RBD-SpyVLP is thermostable and can be lyophilised without losing immunogenicity, to facilitate global distribution and reduce cold-chain dependence. The data suggests that RBD-SpyVLP provides strong potential to address clinical and logistic challenges of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Tiong Kit Tan
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
| | - Pramila Rijal
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Rolle Rahikainen
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Anthony H Keeble
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Lisa Schimanski
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Saira Hussain
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Jack W P Hayes
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Jane C Edwards
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | | | - Miriam Pedrera
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Nazia Thakur
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | | | - Holly Shelton
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Anna Ludi
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | - Clare Browning
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | - Dagmara Bialy
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Sushant Bhat
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | - Philippa Hollinghurst
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
- Department of Microbial Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Matthew Tully
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Katy Moffat
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Chris Chiu
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Ryan Waters
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Ashley Gray
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Mehreen Azhar
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | - Joseph Newman
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Amin S Asfor
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Alison Burman
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | - John A Hammond
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Elma Tchilian
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | - Dalan Bailey
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | | | - Simon P Graham
- The Pirbright Institute, Ash Road, Pirbright, GU24 0NF, UK
| | - Helen M E Duyvesteyn
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Headington, Oxford, UK
| | - Tomas Malinauskas
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Headington, Oxford, UK
| | - Jiandong Huo
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Headington, Oxford, UK
- Rutherford Appleton Laboratory, Protein Production UK, Research Complex at Harwell, and Rosalind Franklin Institute, Harwell, Didcot, OX11 0FA, UK
| | - Julia A Tree
- National Infection Service, Public Health England, Porton Down, Salisbury, SP4 0JG, UK
| | - Karen R Buttigieg
- National Infection Service, Public Health England, Porton Down, Salisbury, SP4 0JG, UK
| | - Raymond J Owens
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Headington, Oxford, UK
- Rutherford Appleton Laboratory, Protein Production UK, Research Complex at Harwell, and Rosalind Franklin Institute, Harwell, Didcot, OX11 0FA, UK
| | - Miles W Carroll
- National Infection Service, Public Health England, Porton Down, Salisbury, SP4 0JG, UK
- Nuffield Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Rodney S Daniels
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - John W McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - David I Stuart
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, The Wellcome Centre for Human Genetics, Headington, Oxford, UK
- Diamond Light Source Ltd, Harwell Science & Innovation Campus, Didcot, UK
| | - Kuan-Ying A Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mark Howarth
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| | - Alain R Townsend
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK.
| |
Collapse
|
18
|
Ng KW, Faulkner N, Cornish GH, Rosa A, Harvey R, Hussain S, Ulferts R, Earl C, Wrobel AG, Benton DJ, Roustan C, Bolland W, Thompson R, Agua-Doce A, Hobson P, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Thomson K, Sanchez E, Shin GY, Spyer MJ, Joshi D, O'Reilly N, Walker PA, Kjaer S, Riddell A, Moore C, Jebson BR, Wilkinson M, Marshall LR, Rosser EC, Radziszewska A, Peckham H, Ciurtin C, Wedderburn LR, Beale R, Swanton C, Gandhi S, Stockinger B, McCauley J, Gamblin SJ, McCoy LE, Cherepanov P, Nastouli E, Kassiotis G. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science 2020; 370:1339-1343. [PMID: 33159009 DOI: 10.1101/2020.05.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/29/2020] [Indexed: 05/20/2023]
Abstract
Zoonotic introduction of novel coronaviruses may encounter preexisting immunity in humans. Using diverse assays for antibodies recognizing SARS-CoV-2 proteins, we detected preexisting humoral immunity. SARS-CoV-2 spike glycoprotein (S)-reactive antibodies were detectable using a flow cytometry-based method in SARS-CoV-2-uninfected individuals and were particularly prevalent in children and adolescents. They were predominantly of the immunoglobulin G (IgG) class and targeted the S2 subunit. By contrast, SARS-CoV-2 infection induced higher titers of SARS-CoV-2 S-reactive IgG antibodies targeting both the S1 and S2 subunits, and concomitant IgM and IgA antibodies, lasting throughout the observation period. SARS-CoV-2-uninfected donor sera exhibited specific neutralizing activity against SARS-CoV-2 and SARS-CoV-2 S pseudotypes. Distinguishing preexisting and de novo immunity will be critical for our understanding of susceptibility to and the natural course of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Kevin W Ng
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | - Nikhil Faulkner
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Annachiara Rosa
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, London NW1 1AT, UK
| | - Saira Hussain
- Worldwide Influenza Centre, The Francis Crick Institute, London NW1 1AT, UK
| | - Rachel Ulferts
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Christopher Earl
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Antoni G Wrobel
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Donald J Benton
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Chloe Roustan
- Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - William Bolland
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | - Rachael Thompson
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | - Ana Agua-Doce
- Flow Cytometry STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Philip Hobson
- Flow Cytometry STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Judith Heaney
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Hannah Rickman
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | | | - Catherine F Houlihan
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK
| | - Kirsty Thomson
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Emilie Sanchez
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Gee Yen Shin
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Moira J Spyer
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
- Department of Population, Policy and Practice, Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Dhira Joshi
- Peptide Chemistry, The Francis Crick Institute, London NW1 1AT, UK
| | - Nicola O'Reilly
- Peptide Chemistry, The Francis Crick Institute, London NW1 1AT, UK
| | - Philip A Walker
- Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Svend Kjaer
- Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Andrew Riddell
- Flow Cytometry STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Catherine Moore
- Public Health Wales, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Bethany R Jebson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Meredyth Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Lucy R Marshall
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Sonia Gandhi
- Neurodegeneration Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | | | - John McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, London NW1 1AT, UK
| | - Steve J Gamblin
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Laura E McCoy
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK.
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| | - Eleni Nastouli
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK.
- Department of Population, Policy and Practice, Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK.
- Department of Medicine, Faculty of Medicine, Imperial College London, London W2 1PG, UK
| |
Collapse
|
19
|
Ng KW, Faulkner N, Cornish GH, Rosa A, Harvey R, Hussain S, Ulferts R, Earl C, Wrobel AG, Benton DJ, Roustan C, Bolland W, Thompson R, Agua-Doce A, Hobson P, Heaney J, Rickman H, Paraskevopoulou S, Houlihan CF, Thomson K, Sanchez E, Shin GY, Spyer MJ, Joshi D, O'Reilly N, Walker PA, Kjaer S, Riddell A, Moore C, Jebson BR, Wilkinson M, Marshall LR, Rosser EC, Radziszewska A, Peckham H, Ciurtin C, Wedderburn LR, Beale R, Swanton C, Gandhi S, Stockinger B, McCauley J, Gamblin SJ, McCoy LE, Cherepanov P, Nastouli E, Kassiotis G. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science 2020; 370:1339-1343. [PMID: 33159009 PMCID: PMC7857411 DOI: 10.1126/science.abe1107] [Citation(s) in RCA: 610] [Impact Index Per Article: 152.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022]
Abstract
Zoonotic introduction of novel coronaviruses may encounter preexisting immunity in humans. Using diverse assays for antibodies recognizing SARS-CoV-2 proteins, we detected preexisting humoral immunity. SARS-CoV-2 spike glycoprotein (S)-reactive antibodies were detectable using a flow cytometry-based method in SARS-CoV-2-uninfected individuals and were particularly prevalent in children and adolescents. They were predominantly of the immunoglobulin G (IgG) class and targeted the S2 subunit. By contrast, SARS-CoV-2 infection induced higher titers of SARS-CoV-2 S-reactive IgG antibodies targeting both the S1 and S2 subunits, and concomitant IgM and IgA antibodies, lasting throughout the observation period. SARS-CoV-2-uninfected donor sera exhibited specific neutralizing activity against SARS-CoV-2 and SARS-CoV-2 S pseudotypes. Distinguishing preexisting and de novo immunity will be critical for our understanding of susceptibility to and the natural course of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Kevin W Ng
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | - Nikhil Faulkner
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Annachiara Rosa
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, London NW1 1AT, UK
| | - Saira Hussain
- Worldwide Influenza Centre, The Francis Crick Institute, London NW1 1AT, UK
| | - Rachel Ulferts
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Christopher Earl
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Antoni G Wrobel
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Donald J Benton
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Chloe Roustan
- Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - William Bolland
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | - Rachael Thompson
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK
| | - Ana Agua-Doce
- Flow Cytometry STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Philip Hobson
- Flow Cytometry STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Judith Heaney
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Hannah Rickman
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | | | - Catherine F Houlihan
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK
| | - Kirsty Thomson
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Emilie Sanchez
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Gee Yen Shin
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
| | - Moira J Spyer
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK
- Department of Population, Policy and Practice, Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Dhira Joshi
- Peptide Chemistry, The Francis Crick Institute, London NW1 1AT, UK
| | - Nicola O'Reilly
- Peptide Chemistry, The Francis Crick Institute, London NW1 1AT, UK
| | - Philip A Walker
- Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Svend Kjaer
- Structural Biology STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Andrew Riddell
- Flow Cytometry STP, The Francis Crick Institute, London NW1 1AT, UK
| | - Catherine Moore
- Public Health Wales, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Bethany R Jebson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Meredyth Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Lucy R Marshall
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- Centre for Rheumatology Research, Division of Medicine, UCL, London, WC1E 6BT, UK
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, Great Ormond Street Hospital (GOSH), London WC1N 3JH, UK
- UCL Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Sonia Gandhi
- Neurodegeneration Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | | | - John McCauley
- Worldwide Influenza Centre, The Francis Crick Institute, London NW1 1AT, UK
| | - Steve J Gamblin
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Laura E McCoy
- Division of Infection and Immunity, University College London (UCL), London WC1E 6BT, UK.
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| | - Eleni Nastouli
- University College London Hospitals (UCLH) NHS Trust, London NW1 2BU, UK.
- Department of Population, Policy and Practice, Great Ormond Street Institute for Child Health (ICH), UCL, London WC1N 1EH, UK
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, London NW1 1AT, UK.
- Department of Medicine, Faculty of Medicine, Imperial College London, London W2 1PG, UK
| |
Collapse
|
20
|
Antibody-mediated disruption of the SARS-CoV-2 spike glycoprotein. Nat Commun 2020; 11:5337. [PMID: 33087721 PMCID: PMC7577971 DOI: 10.1038/s41467-020-19146-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/22/2020] [Indexed: 01/17/2023] Open
Abstract
The CR3022 antibody, selected from a group of SARS-CoV monoclonal antibodies for its ability to cross-react with SARS-CoV-2, has been examined for its ability to bind to the ectodomain of the SARS-CoV-2 spike glycoprotein. Using cryo-electron microscopy we show that antibody binding requires rearrangements in the S1 domain that result in dissociation of the spike. Here, the authors characterise the binding and neutralisation properties of CR3022, a neutralising Ab isolated from a convalescent SARS patient, against SARS-CoV-2 spike trimers and show using Cryo-EM the disruption of SARS-CoV-2 spike by CR3022 Fab.
Collapse
|
21
|
Wang Y, Lv Y, Niu X, Dong J, Feng P, Li Q, Xu W, Li J, Li C, Li J, Luo J, Li Z, Liu Y, Tan YJ, Pan W, Chen L. L226Q Mutation on Influenza H7N9 Virus Hemagglutinin Increases Receptor-Binding Avidity and Leads to Biased Antigenicity Evaluation. J Virol 2020; 94:e00667-20. [PMID: 32796071 PMCID: PMC7527056 DOI: 10.1128/jvi.00667-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/27/2020] [Indexed: 11/20/2022] Open
Abstract
Since the first outbreak in 2013, the influenza A (H7N9) virus has continued emerging and has caused over five epidemic waves. Suspected antigenic changes of the H7N9 virus based on hemagglutination inhibition (HI) assay during the fifth outbreak have prompted the update of H7N9 candidate vaccine viruses (CVVs). In this study, we comprehensively compared the serological cross-reactivities induced by the hemagglutinins (HAs) of the earlier CVV A/Anhui/1/2013 (H7/AH13) and the updated A/Guangdong/17SF003/2016 (H7/GD16). We found that although H7/GD16 showed poor HI cross-reactivity to immune sera from mice and rhesus macaques vaccinated with either H7/AH13 or H7/GD16, the cross-reactive neutralizing antibodies between H7/AH13 and H7/GD16 were comparably high. Passive transfer of H7/AH13 immune sera also provided complete protection against the lethal challenge of H7N9/GD16 virus in mice. Analysis of amino acid mutations in the HAs between H7/AH13 and H7/GD16 revealed that L226Q substitution increases the HA binding avidity to sialic acid receptors on red blood cells, leading to decreased HI titers against viruses containing HA Q226 and thus resulting in a biased antigenic evaluation based on HI assay. These results suggest that amino acids located in the receptor-binding site could mislead the evaluation of antigenic variation by solely impacting the receptor-binding avidity to red blood cells without genuine contribution to antigenic drift. Our study highlighted that viral receptor-binding avidity and combination of multiple serological assays should be taken into consideration in evaluating and selecting a candidate vaccine virus of H7N9 and other subtypes of influenza viruses.IMPORTANCE The HI assay is a standard method for profiling the antigenic characterization of influenza viruses. Suspected antigenic changes based on HI divergency in H7N9 viruses during the 2016-2017 wave prompted the recommendation of new H7N9 candidate vaccine viruses (CVVs). In this study, we found that the L226Q substitution in HA of A/Guangdong/17SF003/2016 (H7/GD16) increased the viral receptor-binding avidity to red blood cells with no impact on the antigenicity of H7N9 virus. Although immune sera raised by an earlier vaccine strain (H7/AH13) showed poor HI titers against H7/GD16, the H7/AH13 immune sera had potent cross-neutralizing antibody titers against H7/GD16 and could provide complete passive protection against H7N9/GD16 virus challenge in mice. Our study highlights that receptor-binding avidity might lead to biased antigenic evaluation by using the HI assay. Other serological assays, such as the microneutralization (MN) assay, should be considered a complementary indicator for analysis of antigenic variation and selection of influenza CVVs.
Collapse
Affiliation(s)
- Yang Wang
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yunhua Lv
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuefeng Niu
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ji Dong
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pei Feng
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qinming Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Xu
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiashun Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chufang Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiahui Li
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jia Luo
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhixia Li
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yichu Liu
- Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yee-Joo Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore, Singapore
| | - Weiqi Pan
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling Chen
- State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
22
|
Comparability of Titers of Antibodies against Seasonal Influenza Virus Strains as Determined by Hemagglutination Inhibition and Microneutralization Assays. J Clin Microbiol 2020; 58:JCM.00750-20. [PMID: 32493784 PMCID: PMC7448638 DOI: 10.1128/jcm.00750-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/22/2020] [Indexed: 11/20/2022] Open
Abstract
We compared titers of antibodies against A/H1N1, A/H3N2, and B influenza virus strains collected pre- and postvaccination using hemagglutination inhibition (HI) and microneutralization (MN) assays and data from two vaccine trials: study 1, performed with a cell-grown trivalent influenza vaccine (TIVc) using cell-grown target virus in both assays, and study 2, performed with an egg-grown adjuvanted quadrivalent influenza vaccine (aQIVe) using egg-grown target virus. The relationships between HI- and MN-derived log-transformed titers were examined using different statistical techniques. We compared titers of antibodies against A/H1N1, A/H3N2, and B influenza virus strains collected pre- and postvaccination using hemagglutination inhibition (HI) and microneutralization (MN) assays and data from two vaccine trials: study 1, performed with a cell-grown trivalent influenza vaccine (TIVc) using cell-grown target virus in both assays, and study 2, performed with an egg-grown adjuvanted quadrivalent influenza vaccine (aQIVe) using egg-grown target virus. The relationships between HI- and MN-derived log-transformed titers were examined using different statistical techniques. Deming regression analyses showed point estimates for slopes generally close to 1 across studies and strains. The slope of regression was closest to 1 for A/H3N2 strain when either cell- or egg-grown viral target virus was used. Bland-Altman plots indicated a very small percentage of results outside 2 and 3 standard deviations. The magnitudes and directions of differences between titers in the two assays varied by study and strain. Mean differences favored the MN assay for A/H1N1 and B strains in study 1, whereas the titers determined by HI were higher than those determined by MN against the A/H3N2 strain. In study 2, mean differences favored the MN assay for A/H3N2 and B strains. Overall, the directions and magnitudes of the mean differences were similar between the two vaccines. The concordance correlation coefficient values ranged from 0.74 (A/H1N1 strain, study 1) to 0.97 (A/H3N2 strain, study 1). The comparative analysis demonstrates an overall strong positive correlation between the HI and MN assays. These data support the use of the MN assay to quantify the immune response of influenza vaccines in clinical studies, particularly for the A/H3N2 strain.
Collapse
|
23
|
Ng TWY, Perera RAPM, Fang VJ, Yau EM, Peiris JSM, Tam YH, Cowling BJ. The Effect of Influenza Vaccination History on Changes in Hemagglutination Inhibition Titers After Receipt of the 2015-2016 Influenza Vaccine in Older Adults in Hong Kong. J Infect Dis 2020; 221:33-41. [PMID: 31282541 DOI: 10.1093/infdis/jiz327] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 06/25/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Immune responses to influenza vaccination can be weaker in older adults than in other age groups. We hypothesized that antibody responses would be particularly weak among repeat vaccinees when the current and prior season vaccine components are the same. METHODS An observational study was conducted among 827 older adults (aged ≥75 years) in Hong Kong. Serum samples were collected immediately before and 1 month after receipt of the 2015-2016 quadrivalent inactivated influenza vaccine. We measured antibody titers with the hemagglutination inhibition assay and compared the mean fold rise from prevaccination to postvaccination titers and the proportions with postvaccination titers ≥40 or ≥160. RESULTS Participants who reported receipt of vaccination during either of the previous 2 years had a lower mean fold rise against all strains than with those who did not. Mean fold rises for A(H3N2) and B/Yamagata were particularly weak after repeated vaccination with the same vaccine strain, but we did not generally find significant differences in the proportions of participants with postvaccination titers ≥40 and ≥160. CONCLUSIONS Overall, we found that reduced antibody responses in repeat vaccinees were particularly reduced among older adults who had received vaccination against the same strains in preceding years.
Collapse
Affiliation(s)
- Tiffany W Y Ng
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ranawaka A P M Perera
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Vicky J Fang
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Emily M Yau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - J S Malik Peiris
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yat Hung Tam
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
24
|
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
|
25
|
Jorquera PA, Mishin VP, Chesnokov A, Nguyen HT, Mann B, Garten R, Barnes J, Hodges E, De La Cruz J, Xu X, Katz J, Wentworth DE, Gubareva LV. Insights into the antigenic advancement of influenza A(H3N2) viruses, 2011-2018. Sci Rep 2019; 9:2676. [PMID: 30804469 PMCID: PMC6389938 DOI: 10.1038/s41598-019-39276-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/11/2019] [Indexed: 11/12/2022] Open
Abstract
Influenza A(H3N2) viruses evade human immunity primarily by acquiring antigenic changes in the haemagglutinin (HA). HA receptor-binding features of contemporary A(H3N2) viruses hinder traditional antigenic characterization using haemagglutination inhibition and promote selection of HA mutants. Thus, alternative approaches are needed to reliably assess antigenic relatedness between circulating viruses and vaccines. We developed a high content imaging-based neutralization test (HINT) to reduce antigenic mischaracterization resulting from virus adaptation to cell culture. Ferret reference antisera were raised using clinical specimens containing viruses representing recent vaccine strains. Analysis of viruses circulating during 2011-2018 showed that gain of an N158-linked glycosylation in HA was a molecular determinant of antigenic distancing between A/Hong Kong/4801/2014-like (clade 3C.2a) and A/Texas/50/2012-like viruses (clade 3C.1), while multiple evolutionary HA F193S substitution were linked to antigenic distancing from A/Switzerland/97152963/2013-like (clade 3C.3a) and further antigenic distancing from A/Texas/50/2012-like viruses. Additionally, a few viruses carrying HA T135K and/or I192T showed reduced neutralization by A/Hong Kong/4801/2014-like antiserum. Notably, this technique elucidated the antigenic characteristics of clinical specimens, enabling direct characterization of viruses produced in vivo, and eliminating in vitro culture, which rapidly alters the genotype/phenotype. HINT is a valuable new antigenic analysis tool for vaccine strain selection.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Antigens, Viral/immunology
- Ferrets/immunology
- Ferrets/virology
- Glycosylation
- Hemagglutination Inhibition Tests/methods
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Humans
- Immune Sera/immunology
- Influenza A Virus, H3N2 Subtype/classification
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/physiology
- Influenza Vaccines/immunology
- Influenza, Human/diagnosis
- Influenza, Human/immunology
- Influenza, Human/virology
- Neutralization Tests/methods
- Phylogeny
Collapse
Affiliation(s)
- Patricia A Jorquera
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- CNI Advantage, LLC. 17 Executive Park Dr NE, Atlanta, GA, 30329, USA
| | - Vasiliy P Mishin
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Anton Chesnokov
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Ha T Nguyen
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- Battelle Memorial Institute, 2987 Clairmont Rd, Suite 450, Atlanta, GA, 30329, USA
| | - Brian Mann
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- Battelle Memorial Institute, 2987 Clairmont Rd, Suite 450, Atlanta, GA, 30329, USA
| | - Rebecca Garten
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - John Barnes
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Erin Hodges
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- CNI Advantage, LLC. 17 Executive Park Dr NE, Atlanta, GA, 30329, USA
| | - Juan De La Cruz
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
- Battelle Memorial Institute, 2987 Clairmont Rd, Suite 450, Atlanta, GA, 30329, USA
| | - Xiyan Xu
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Jackie Katz
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - David E Wentworth
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA
| | - Larisa V Gubareva
- Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road, Atlanta, GA, 30329, USA.
| |
Collapse
|
26
|
Beer K, Dai M, Howell S, Rijal P, Townsend AR, Lin Y, Wharton SA, Daniels RS, McCauley JW. Characterization of neutralizing epitopes in antigenic site B of recently circulating influenza A(H3N2) viruses. J Gen Virol 2018; 99:1001-1011. [PMID: 29944110 PMCID: PMC6171714 DOI: 10.1099/jgv.0.001101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Influenza A(H3N2) viruses are associated with outbreaks worldwide and can cause disease with severe complications. The impact can be reduced by vaccination, which induces neutralizing antibodies that mainly target the haemagglutinin glycoprotein (HA). In this study we generated neutralizing mouse monoclonal antibodies (mAbs) against A/Victoria/361/2011 and identified their epitopes by generating and sequencing escape viruses. The epitopes are located in antigenic site B, which is near the receptor-binding site and is immunodominant in humans. Amino acid (aa) substitutions at positions 156, 158, 159, 189, 190 and 193 in antigenic site B led to reduced ability of mAbs to block receptor-binding. The majority of A(H3N2) viruses that have been circulating since 2014 are antigenically distinct from previous A(H3N2) viruses. The neutralization-sensitive epitopes in antigenic site B of currently circulating viruses were examined with these mAbs. We found that clade 3C.2a viruses, possessing an additional potential glycosylation site at HA1 position N158, were poorly recognized by some of the mAbs, but other residues, notably at position 159, also affected antibody binding. Through a mass spectrometric (MS) analysis of HA, the glycosylated sites of HA1 were established and we determined that residue 158 of HA1 was glycosylated and so modified a neutralization-sensitive epitope. Understanding and monitoring individual epitopes is likely to improve vaccine strain selection.
Collapse
Affiliation(s)
- Kerstin Beer
- 1Crick Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Mian Dai
- 1Crick Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Steven Howell
- 2Mass Spectrometry and Proteomics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Pramila Rijal
- 3MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Alain R Townsend
- 3MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Yipu Lin
- 1Crick Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Stephen A Wharton
- 1Crick Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rodney S Daniels
- 1Crick Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - John W McCauley
- 1Crick Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
27
|
Tilmanis D, van Baalen C, Oh DY, Rossignol JF, Hurt AC. The susceptibility of circulating human influenza viruses to tizoxanide, the active metabolite of nitazoxanide. Antiviral Res 2017; 147:142-148. [DOI: 10.1016/j.antiviral.2017.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/22/2017] [Accepted: 10/02/2017] [Indexed: 10/18/2022]
|
28
|
Mögling R, Richard MJ, Vliet SVD, Beek RV, Schrauwen EJA, Spronken MI, Rimmelzwaan GF, Fouchier RAM. Neuraminidase-mediated haemagglutination of recent human influenza A(H3N2) viruses is determined by arginine 150 flanking the neuraminidase catalytic site. J Gen Virol 2017; 98:1274-1281. [PMID: 28612701 DOI: 10.1099/jgv.0.000809] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Over the last decade, an increasing proportion of circulating human influenza A(H3N2) viruses exhibited haemagglutination activity that was sensitive to neuraminidase inhibitors. This change in haemagglutination as compared to older circulating A(H3N2) viruses prompted an investigation of the underlying molecular basis. Recent human influenza A(H3N2) viruses were found to agglutinate turkey erythrocytes in a manner that could be blocked with either oseltamivir or neuraminidase-specific antisera, indicating that agglutination was driven by neuraminidase, with a low or negligible contribution of haemagglutinin. Using representative virus recombinants it was shown that the haemagglutinin of a recent A(H3N2) virus indeed had decreased activity to agglutinate turkey erythrocytes, while its neuraminidase displayed increased haemagglutinating activity. Viruses with chimeric and mutant neuraminidases were used to identify the amino acid substitution histidine to arginine at position 150 flanking the neuraminidase catalytic site as the determinant of this neuraminidase-mediated haemagglutination. An analysis of publicly available neuraminidase gene sequences showed that viruses with histidine at position 150 were rapidly replaced by viruses with arginine at this position between 2005 and 2008, in agreement with the phenotypic data. As a consequence of neuraminidase-mediated haemagglutination of recent A(H3N2) viruses and poor haemagglutination via haemagglutinin, haemagglutination inhibition assays with A(H3N2) antisera are no longer useful to characterize the antigenic properties of the haemagglutinin of these viruses for vaccine strain selection purposes. Continuous monitoring of the evolution of these viruses and potential consequences for vaccine strain selection remains important.
Collapse
Affiliation(s)
- Ramona Mögling
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - Ruud van Beek
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Eefje J A Schrauwen
- Laboratory for Microbiology and Infection Control, Amphia Hospital, Breda, The Netherlands
| | | | | | - Ron A M Fouchier
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Hollý J, Fogelová M, Jakubcová L, Tomčíková K, Vozárová M, Varečková E, Kostolanský F. Comparison of infectious influenza A virus quantification methods employing immuno-staining. J Virol Methods 2017; 247:107-113. [PMID: 28610903 DOI: 10.1016/j.jviromet.2017.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/09/2017] [Accepted: 06/10/2017] [Indexed: 11/16/2022]
Abstract
Infections caused by highly variable influenza A viruses (IAVs) pose perpetual threat to humans as well as to animals. Their surveillance requires reliable methods for their qualitative and quantitative analysis. The most frequently utilized quantification method is the titration by plaque assay or 50% tissue culture infectious dose estimation by TCID50. However, both methods are time-consuming. Moreover, some IAV strains form hardly visible plaques, and the evaluation of TCID50 is subjective. Employment of immuno-staining into the classic protocols for plaque assay or TCID50 assay enables to avoid these problems and moreover, shorten the time needed for reliable infectious virus quantification. Results obtained by these two alternatives of classic virus titration methods were compared to the newer rapid culture assay (RCA), where titration endpoint of infectious virus was estimated microscopically based on the immuno-staining of infected cells. In our analysis of compared methods, five different IAV strains of H1, H3 and H5 subtypes were used and results were statistically evaluated. We conclude that the RCA proved to be at least as reliable in assessment of infectious viral titer as plaque assay and TCID50, considering the employed immuno-staining.
Collapse
Affiliation(s)
- Jaroslav Hollý
- Biomedical Research Center, Institute of Virology, Department of Orthomyxoviruses, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovak Republic
| | - Margaréta Fogelová
- Biomedical Research Center, Institute of Virology, Department of Orthomyxoviruses, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovak Republic
| | - Lucia Jakubcová
- Biomedical Research Center, Institute of Virology, Department of Orthomyxoviruses, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovak Republic
| | - Karolína Tomčíková
- Biomedical Research Center, Institute of Virology, Department of Orthomyxoviruses, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovak Republic
| | - Mária Vozárová
- Biomedical Research Center, Institute of Virology, Department of Orthomyxoviruses, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovak Republic
| | - Eva Varečková
- Biomedical Research Center, Institute of Virology, Department of Orthomyxoviruses, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovak Republic.
| | - František Kostolanský
- Biomedical Research Center, Institute of Virology, Department of Orthomyxoviruses, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05, Bratislava, Slovak Republic
| |
Collapse
|
30
|
Lin Y, Wharton SA, Whittaker L, Dai M, Ermetal B, Lo J, Pontoriero A, Baumeister E, Daniels RS, McCauley JW. The characteristics and antigenic properties of recently emerged subclade 3C.3a and 3C.2a human influenza A(H3N2) viruses passaged in MDCK cells. Influenza Other Respir Viruses 2017; 11:263-274. [PMID: 28164446 PMCID: PMC5410720 DOI: 10.1111/irv.12447] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2017] [Indexed: 11/28/2022] Open
Abstract
Background Two new subclades of influenza A(H3N2) viruses became prominent during the 2014‐2015 Northern Hemisphere influenza season. The HA glycoproteins of these viruses showed sequence changes previously associated with alterations in receptor‐binding properties. To address how these changes influence virus propagation, viruses were isolated and propagated in conventional MDCK cells and MDCK‐SIAT1 cells, cells with enhanced expression of the human receptor for the virus, and analysed at each passage. Methods Gene sequence analysis was undertaken as virus was passaged in conventional MDCK cells and MDCK‐SIAT1 cells. Alterations in receptor recognition associated with passage of virus were examined by haemagglutination assays using red blood cells from guinea pigs, turkeys and humans. Microneutralisation assays were performed to determine how passage‐acquired amino acid substitutions and polymorphisms affected virus antigenicity. Results Viruses were able to infect MDCK‐SIAT1 cells more efficiently than conventional MDCK cells. Viruses of both the 3C.2a and 3C.3a subclades showed greater sequence change on passage in conventional MDCK cells than in MDCK‐SIAT1 cells, with amino acid substitutions being seen in both HA and NA glycoproteins. However, virus passage in MDCK‐SIAT1 cells at low inoculum dilutions showed reducing infectivity on continued passage. Conclusions Current H3N2 viruses should be cultured in the MDCK‐SIAT1 cell line to maintain faithful replication of the virus, and at an appropriate multiplicity of infection to retain infectivity.
Collapse
Affiliation(s)
- Yipu Lin
- The Francis Crick Institute, London, UK
| | | | | | - Mian Dai
- The Francis Crick Institute, London, UK
| | | | - Janice Lo
- Centre for Health Protection, Department of Health, Hong Kong Special Administrative Region, China
| | - Andrea Pontoriero
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr.Carlos G.Malbran", Buenos Aires, Argentina
| | - Elsa Baumeister
- Instituto Nacional de Enfermedades Infecciosas-ANLIS "Dr.Carlos G.Malbran", Buenos Aires, Argentina
| | | | | |
Collapse
|
31
|
van Baalen CA, Jeeninga RE, Penders GH, van Gent B, van Beek R, Koopmans MP, Rimmelzwaan GF. ViroSpot microneutralization assay for antigenic characterization of human influenza viruses. Vaccine 2017; 35:46-52. [DOI: 10.1016/j.vaccine.2016.11.060] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 12/21/2022]
|
32
|
Abstract
The micro-neutralization (MN) assay is a standard technique for measuring the infectivity of the influenza virus and the inhibition of virus replication. In this study, we present the protocol of an imaging-based MN assay to quantify the true antigenic relationships between viruses. Unlike typical plaque reduction assays that rely on visible plaques, this assay quantitates the entire infected cell population of each well. The protocol matches the virus type or subtype with the selection of cell lines to achieve maximum infectivity, which enhances sample contrast during imaging and image processing. The introduction of quantitative titration defines the amount of input viruses of neutralization and enables the results from different experiments to be comparable. The imaging setup with a flatbed scanner and free downloadable software makes the approach high throughput, cost effective, user friendly, and easy to deploy in most laboratories. Our study demonstrates that the improved MN assay works well with the current circulating influenza A(H1N1)pdm09, A(H3N2), and B viruses, without being significantly influenced by amino acid substitutions in the neuraminidase (NA) of A(H3N2) viruses. It is particularly useful for the characterization of viruses that either grow to low HA titer and/or undergo an abortive infection resulting in an inability to form plaques in cultured cells.
Collapse
Affiliation(s)
- Yipu Lin
- Mill Hill Laboratory, The Francis Crick Institute
| | - Yan Gu
- Mill Hill Laboratory, The Francis Crick Institute;
| | | |
Collapse
|
33
|
Prediction of influenza B vaccine effectiveness from sequence data. Vaccine 2016; 34:4610-4617. [PMID: 27473305 DOI: 10.1016/j.vaccine.2016.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/01/2016] [Accepted: 07/12/2016] [Indexed: 11/22/2022]
Abstract
Influenza is a contagious respiratory illness that causes significant human morbidity and mortality, affecting 5-15% of the population in a typical epidemic season. Human influenza epidemics are caused by types A and B, with roughly 25% of human cases due to influenza B. Influenza B is a single-stranded RNA virus with a high mutation rate, and both prior immune history and vaccination put significant pressure on the virus to evolve. Due to the high rate of viral evolution, the influenza B vaccine component of the annual influenza vaccine is updated, roughly every other year in recent years. To predict when an update to the vaccine is needed, an estimate of expected vaccine effectiveness against a range of viral strains is required. We here introduce a method to measure antigenic distance between the influenza B vaccine and circulating viral strains. The measure correlates well with effectiveness of the influenza B component of the annual vaccine in humans between 1979 and 2014. We discuss how this measure of antigenic distance may be used in the context of annual influenza vaccine design and prediction of vaccine effectiveness.
Collapse
|