99 results on '"P Supasa"'
Search Results
2. The SARS-CoV-2 neutralizing antibody response to SD1 and its evasion by BA.2.86
- Author
-
Zhou, Daming, Supasa, Piyada, Liu, Chang, Dijokaite-Guraliuc, Aiste, Duyvesteyn, Helen M. E., Selvaraj, Muneeswaran, Mentzer, Alexander J., Das, Raksha, Dejnirattisai, Wanwisa, Temperton, Nigel, Klenerman, Paul, Dunachie, Susanna J., Fry, Elizabeth E., Mongkolsapaya, Juthathip, Ren, Jingshan, Stuart, David I., and Screaton, Gavin R.
- Published
- 2024
- Full Text
- View/download PDF
3. Omicron infection following vaccination enhances a broad spectrum of immune responses dependent on infection history
- Author
-
Hornsby, Hailey, Nicols, Alexander R., Longet, Stephanie, Liu, Chang, Tomic, Adriana, Angyal, Adrienn, Kronsteiner, Barbara, Tyerman, Jessica K., Tipton, Tom, Zhang, Peijun, Gallis, Marta, Supasa, Piyada, Selvaraj, Muneeswaran, Abraham, Priyanka, Neale, Isabel, Ali, Mohammad, Barratt, Natalie A., Nell, Jeremy M., Gustafsson, Lotta, Strickland, Scarlett, Grouneva, Irina, Rostron, Timothy, Moore, Shona C., Hering, Luisa M., Dobson, Susan L., Bibi, Sagida, Mongkolsapaya, Juthathip, Lambe, Teresa, Wootton, Dan, Hall, Victoria, Hopkins, Susan, Dong, Tao, Barnes, Eleanor, Screaton, Gavin, Richter, Alex, Turtle, Lance, Rowland-Jones, Sarah L., Carroll, Miles, Duncan, Christopher J. A., Klenerman, Paul, Dunachie, Susanna J., Payne, Rebecca P., and de Silva, Thushan I.
- Published
- 2023
- Full Text
- View/download PDF
4. Antigenic characterization of SARS-CoV-2 Omicron subvariant BA.4.6
- Author
-
Dijokaite-Guraliuc, Aiste, Das, Raksha, Nutalai, Rungtiwa, Zhou, Daming, Mentzer, Alexander J., Liu, Chang, Supasa, Piyada, Dunachie, Susanna J., Lambe, Teresa, Fry, Elizabeth E., Mongkolsapaya, Juthathip, Ren, Jingshan, Huo, Jiandong, Stuart, David I., and Screaton, Gavin R.
- Published
- 2022
- Full Text
- View/download PDF
5. Structural basis for the neutralization of SARS-CoV-2 by an antibody from a convalescent patient
- Author
-
Zhou, Daming, Duyvesteyn, Helen M. E., Chen, Cheng-Pin, Huang, Chung-Guei, Chen, Ting-Hua, Shih, Shin-Ru, Lin, Yi-Chun, Cheng, Chien-Yu, Cheng, Shu-Hsing, Huang, Yhu-Chering, Lin, Tzou-Yien, Ma, Che, Huo, Jiandong, Carrique, Loic, Malinauskas, Tomas, Ruza, Reinis R., Shah, Pranav N. M., Tan, Tiong Kit, Rijal, Pramila, Donat, Robert F., Godwin, Kerry, Buttigieg, Karen R., Tree, Julia A., Radecke, Julika, Paterson, Neil G., Supasa, Piyada, Mongkolsapaya, Juthathip, Screaton, Gavin R., Carroll, Miles W., Gilbert-Jaramillo, Javier, Knight, Michael L., James, William, Owens, Raymond J., Naismith, James H., Townsend, Alain R., Fry, Elizabeth E., Zhao, Yuguang, Ren, Jingshan, Stuart, David I., and Huang, Kuan-Ying A.
- Published
- 2020
- Full Text
- View/download PDF
6. A new class of highly potent, broadly neutralizing antibodies isolated from viremic patients infected with dengue virus
- Author
-
Dejnirattisai, Wanwisa, Wongwiwat, Wiyada, Supasa, Sunpetchuda, Zhang, Xiaokang, Dai, Xinghong, Rouvinski, Alexander, Jumnainsong, Amonrat, Edwards, Carolyn, Quyen, Nguyen Than Ha, Duangchinda, Thaneeya, Grimes, Jonathan M, Tsai, Wen-Yang, Lai, Chih-Yun, Wang, Wei-Kung, Malasit, Prida, Farrar, Jeremy, Simmons, Cameron P, Zhou, Z Hong, Rey, Felix A, Mongkolsapaya, Juthathip, and Screaton, Gavin R
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Immunology ,Prevention ,Infectious Diseases ,Emerging Infectious Diseases ,Vaccine Related ,Immunization ,Rare Diseases ,Vector-Borne Diseases ,Biotechnology ,Biodefense ,Infection ,Good Health and Well Being ,Animals ,Antibodies ,Monoclonal ,Antibodies ,Neutralizing ,Biological Assay ,Cell Line ,Dengue ,Dengue Virus ,Enzyme-Linked Immunosorbent Assay ,Humans ,Immunoblotting ,Viral Envelope Proteins ,Biochemistry and cell biology - Abstract
Dengue is a rapidly emerging, mosquito-borne viral infection, with an estimated 400 million infections occurring annually. To gain insight into dengue immunity, we characterized 145 human monoclonal antibodies (mAbs) and identified a previously unknown epitope, the envelope dimer epitope (EDE), that bridges two envelope protein subunits that make up the 90 repeating dimers on the mature virion. The mAbs to EDE were broadly reactive across the dengue serocomplex and fully neutralized virus produced in either insect cells or primary human cells, with 50% neutralization in the low picomolar range. Our results provide a path to a subunit vaccine against dengue virus and have implications for the design and monitoring of future vaccine trials in which the induction of antibody to the EDE should be prioritized.
- Published
- 2015
7. A haemagglutination test for rapid detection of antibodies to SARS-CoV-2
- Author
-
Townsend, Alain, Rijal, Pramila, Xiao, Julie, Tan, Tiong Kit, Huang, Kuan-Ying A., Schimanski, Lisa, Huo, Jiandong, Gupta, Nimesh, Rahikainen, Rolle, Matthews, Philippa C., Crook, Derrick, Hoosdally, Sarah, Dunachie, Susanna, Barnes, Eleanor, Street, Teresa, Conlon, Christopher P., Frater, John, Arancibia-Cárcamo, Carolina V., Rudkin, Justine, Stoesser, Nicole, Karpe, Fredrik, Neville, Matthew, Ploeg, Rutger, Oliveira, Marta, Roberts, David J., Lamikanra, Abigail A., Tsang, Hoi Pat, Bown, Abbie, Vipond, Richard, Mentzer, Alexander J., Knight, Julian C., Kwok, Andrew J., Screaton, Gavin R., Mongkolsapaya, Juthathip, Dejnirattisai, Wanwisa, Supasa, Piyada, Klenerman, Paul, Dold, Christina, Baillie, J. Kenneth, Moore, Shona C., Openshaw, Peter J. M., Semple, Malcolm G., Turtle, Lance C. W., Ainsworth, Mark, Allcock, Alice, Beer, Sally, Bibi, Sagida, Skelly, Donal, Stafford, Lizzy, Jeffrey, Katie, O’Donnell, Denise, Clutterbuck, Elizabeth, Espinosa, Alexis, Mendoza, Maria, Georgiou, Dominique, Lockett, Teresa, Martinez, Jose, Perez, Elena, Gallardo Sanchez, Veronica, Scozzafava, Giuseppe, Sobrinodiaz, Alberto, Thraves, Hannah, and Joly, Etienne
- Published
- 2021
- Full Text
- View/download PDF
8. Characterization of a potent and highly unusual minimally enhancing antibody directed against dengue virus
- Author
-
Renner, Max, Flanagan, Aleksandra, Dejnirattisai, Wanwisa, Puttikhunt, Chunya, Kasinrerk, Watchara, Supasa, Piyada, Wongwiwat, Wiyada, Chawansuntati, Kriangkrai, Duangchinda, Thaneeya, Cowper, Alison, Midgley, Claire M., Malasit, Prida, Huiskonen, Juha T., Mongkolsapaya, Juthathip, Screaton, Gavin R., and Grimes, Jonathan M.
- Published
- 2018
- Full Text
- View/download PDF
9. Therapeutic and protective efficacy of a dengue antibody against Zika infection in rhesus monkeys
- Author
-
Abbink, Peter, Larocca, Rafael A., Dejnirattisai, Wanwisa, Peterson, Rebecca, Nkolola, Joseph P., Borducchi, Erica N., Supasa, Piyada, Mongkolsapaya, Juthathip, Screaton, Gavin R., and Barouch, Dan H.
- Published
- 2018
- Full Text
- View/download PDF
10. Human antibodies to the dengue virus E-dimer epitope have therapeutic activity against Zika virus infection
- Author
-
Fernandez, Estefania, Dejnirattisai, Wanwisa, Cao, Bin, Scheaffer, Suzanne M, Supasa, Piyada, Wongwiwat, Wiyada, Esakky, Prabagaran, Drury, Andrea, Mongkolsapaya, Juthathip, Moley, Kelle H, Mysorekar, Indira U, Screaton, Gavin R, and Diamond, Michael S
- Published
- 2017
- Full Text
- View/download PDF
11. Author Correction: Human antibodies to the dengue virus E-dimer epitope have therapeutic activity against Zika virus infection
- Author
-
Fernandez, Estefania, Dejnirattisai, Wanwisa, Cao, Bin, Scheaffer, Suzanne M, Supasa, Piyada, Wongwiwat, Wiyada, Esakky, Prabagaran, Drury, Andrea, Mongkolsapaya, Juthathip, Moley, Kelle H, Mysorekar, Indira U, Screaton, Gavin R, and Diamond, Michael S
- Published
- 2020
- Full Text
- View/download PDF
12. Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with zika virus
- Author
-
Dejnirattisai, Wanwisa, Supasa, Piyada, Wongwiwat, Wiyada, Rouvinski, Alexander, Barba-Spaeth, Giovanna, Duangchinda, Thaneeya, Sakuntabhai, Anavaj, Cao-Lormeau, Van-Mai, Malasit, Prida, Rey, Felix A, Mongkolsapaya, Juthathip, and Screaton, Gavin R
- Published
- 2016
- Full Text
- View/download PDF
13. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19
- Author
-
Christina Dold, Mariolina Salio, John Frater, Peter Simmonds, Christopher P. Conlon, J Slon-Campos, Z Yin, David I. Stuart, N F Schwabe, Guido C. Paesen, D Dong, Richard J. Cornall, Paul Thomson, T Lockett, Oliver W. Bayfield, U S Rajapaksa, J W Fry, P Supasa, Jonathan M. Grimes, R Levin, Wanwisa Dejnirattisai, Graham S. Ogg, Dannielle Wellington, Openshaw Pjm., Chen Y-L., Hawkins Dedp., P Zhang, Giorgio Napolitani, Tao Dong, J K Baillie, C Liu, Cesar Lopez-Camacho, Paul Sopp, M A Ansari, Alfred A. Antson, Ker D-S., Persephone Borrow, Susanna Dunachie, Andrew J. McMichael, Juthathip Mongkolsapaya, Yanchun Peng, B Wang, T Rostron, Shona C Moore, Lance Turtle, G Liu, T I de Silva, Krishanthi Subramaniam, Jeremy Ratcliff, Ho L-P., Benedikt M. Kessler, G Kerr, Yuguang Zhao, X Yao, Malcolm G Semple, Gavin R. Screaton, Paul Klenerman, Wayne Paes, R Jing, Alexander J. Mentzer, Y Zhang, Philip J. R. Goulder, Eleanor Barnes, and Julian C. Knight
- Subjects
0301 basic medicine ,Effector ,business.industry ,T cell ,Immunology ,Disease ,Epitope ,Article ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Antigen ,Immunity ,medicine ,Cytotoxic T cell ,Immunology and Allergy ,business ,CD8 ,030215 immunology - Abstract
The development of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines and therapeutics will depend on understanding viral immunity. We studied T cell memory in 42 patients following recovery from COVID-19 (28 with mild disease and 14 with severe disease) and 16 unexposed donors, using interferon-γ-based assays with peptides spanning SARS-CoV-2 except ORF1. The breadth and magnitude of T cell responses were significantly higher in severe as compared with mild cases. Total and spike-specific T cell responses correlated with spike-specific antibody responses. We identified 41 peptides containing CD4+ and/or CD8+ epitopes, including six immunodominant regions. Six optimized CD8+ epitopes were defined, with peptide–MHC pentamer-positive cells displaying the central and effector memory phenotype. In mild cases, higher proportions of SARS-CoV-2-specific CD8+ T cells were observed. The identification of T cell responses associated with milder disease will support an understanding of protective immunity and highlights the potential of including non-spike proteins within future COVID-19 vaccine design. Questions have arisen as to whether patients with severe COVID-19 disease can generate a T cell response against SARS-CoV-2. Tao Dong and colleagues report that convalescent patients with COVID-19 harbor functional memory CD4+ and CD8+ T cells that recognize multiple epitopes that span the viral proteome. CD4+ T cells predominated the memory response in patients with severe disease, whereas higher proportions of CD8+ T cells were found in patients with mild disease.
- Published
- 2022
14. Antibody evasion by the P.1 strain of SARS-CoV-2
- Author
-
Beibei Wang, D. Zhou, Elizabeth E. Fry, Julian C. Knight, Amy Flaxman, Valdinete Alves do Nascimento, Alexander J. Mentzer, E Barnes, Alex Pauvolid-Corrêa, Sarah C. Gilbert, M Bittaye, Mark A. Williams, Hulswit Rjg., Gavin R. Screaton, Sandra Belij-Rammerstorfer, Chang Liu, Andrew J. Pollard, David R. Hall, Tao Dong, David I. Stuart, Christina Dold, Felipe Gomes Naveca, Wanwisa Dejnirattisai, Elizabeth A. Clutterbuck, Paul Klenerman, Neil G. Paterson, Helen M. Ginn, C Fernandes da Costa, R Levin, Jingshan Ren, Duyvesteyn Hme., Thomas S. Walter, Aekkachai Tuekprakhon, Miles W. Carroll, Donal T. Skelly, R Nutalai, Yuguang Zhao, Thomas A. Bowden, D W Crook, S A Costa Clemens, Marilda M. Siqueira, P Supasa, Susanna Dunachie, S Bibi, Teresa Lambe, Paola Cristina Resende, Cesar Lopez-Camacho, J Slon-Campos, Juthathip Mongkolsapaya, and Fabrícia F. Nascimento
- Subjects
medicine.drug_class ,medicine.disease_cause ,Monoclonal antibody ,Immunoglobulin light chain ,Antibodies, Viral ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Neutralization ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,medicine ,Humans ,Binding site ,Neutralizing antibody ,COVID-19 Serotherapy ,030304 developmental biology ,Immune Evasion ,Sequence Deletion ,0303 health sciences ,Mutation ,Vaccines ,Binding Sites ,biology ,SARS-CoV-2 ,Vaccination ,Immunization, Passive ,Antibodies, Monoclonal ,COVID-19 ,Virology ,Antibodies, Neutralizing ,Spike Glycoprotein, Coronavirus ,biology.protein ,Antibody ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Terminating the SARS-CoV-2 pandemic relies upon pan-global vaccination. Current vaccines elicit neutralizing antibody responses to the virus spike derived from early isolates. However, new strains have emerged with multiple mutations: P.1 from Brazil, B.1.351 from South Africa and B.1.1.7 from the UK (12, 10 and 9 changes in the spike respectively). All have mutations in the ACE2 binding site with P.1 and B.1.351 having a virtually identical triplet: E484K, K417N/T and N501Y, which we show confer similar increased affinity for ACE2. We show that, surprisingly, P.1 is significantly less resistant to naturally acquired or vaccine induced antibody responses than B.1.351 suggesting that changes outside the RBD impact neutralisation. Monoclonal antibody 222 neutralises all three variants despite interacting with two of the ACE2 binding site mutations, we explain this through structural analysis and use the 222 light chain to largely restore neutralization potency to a major class of public antibodies., Structural and functional analysis of the P.1 variant of SARS-CoV-2 from Brazil reveals less resistance to antibodies generated from natural infection or vaccination compared to another similar variant, B.1.351. A monoclonal antibody mAb 222 is able to neutralize all three variants (P.1, B.1.351 and B.1.1.7), with its light chain able to restore neutralization potency to broad group of antibodies.
- Published
- 2021
- Full Text
- View/download PDF
15. Reduced neutralization of SARS-CoV-2 B.1.617 by vaccine and convalescent serum
- Author
-
Christina Dold, Miles W. Carroll, C Mason, David Hall, Eleanor Barnes, Cesar Lopez-Camacho, F. Gomes Naveca, Jingshan Ren, M C Nunes, D. Zhou, David I. Stuart, P Goulder, Elizabeth E. Fry, Neil G. Paterson, Shabir A. Madhi, Juthathip Mongkolsapaya, Amy Flaxman, S A Johnson, Teresa Lambe, Valdinete Alves do Nascimento, Sandra Belij-Rammerstorfer, Alexander J. Mentzer, Helen M. Ginn, Tao Dong, Thomas S. Walter, Donal T. Skelly, Paul Klenerman, Z Ditse, Marilda M. Siqueira, Sarah C. Gilbert, Susanna Dunachie, James C. Knight, Alex Pauvolid-Corrêa, P Supasa, Mark A. Williams, Vicky L. Baillie, N Serafin, C Fernandes da Costa, Yuguang Zhao, Andrew J. Pollard, R Nutalai, S Bibi, T G Ritter, Fabrícia F. Nascimento, M Bittaye, Wanwisa Dejnirattisai, Beibei Wang, Chang Liu, Gavin R. Screaton, Elizabeth A. Clutterbuck, Aekkachai Tuekprakhon, Paola Cristina Resende, J Slon-Campos, S A Costa Clemens, Nigel J. Temperton, Duyvesteyn Hme., D W Crook, K Da Silva, and T Malik
- Subjects
Antigen-Antibody Complex ,COVID-19 Vaccines ,medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Antibodies, Viral ,Crystallography, X-Ray ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,Article ,Protein Domains ,Antigen ,Neutralization Tests ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Vero Cells ,COVID-19 Serotherapy ,QR355 ,biology ,SARS-CoV-2 ,Immunization, Passive ,Antibodies, Monoclonal ,COVID-19 ,Antibodies, Neutralizing ,Virology ,Immunization ,Spike Glycoprotein, Coronavirus ,biology.protein ,Vero cell ,Antibody - Abstract
SARS-CoV-2 has undergone progressive change with variants conferring advantage rapidly becoming dominant lineages e.g. B.1.617. With apparent increased transmissibility variant B.1.617.2 has contributed to the current wave of infection ravaging the Indian subcontinent and has been designated a variant of concern in the UK. Here we study the ability of monoclonal antibodies, convalescent and vaccine sera to neutralize B.1.617.1 and B.1.617.2 and complement this with structural analyses of Fab/RBD complexes and map the antigenic space of current variants. Neutralization of both viruses is reduced when compared with ancestral Wuhan related strains but there is no evidence of widespread antibody escape as seen with B.1.351. However, B.1.351 and P.1 sera showed markedly more reduction in neutralization of B.1.617.2 suggesting that individuals previously infected by these variants may be more susceptible to reinfection by B.1.617.2. This observation provides important new insight for immunisation policy with future variant vaccines in non-immune populations., The B.1.617 lineage of SARS-CoV-2, especially the delta strain that is B.1.617.2 has contributed to the wave of infection in the Indian subcontinent. Structural and serological analyses show no evidence of antibody escape but individuals previously infected with either the B.1.351 (beta) and P.1 (gamma) variants are likely more susceptible to reinfection by the delta strain. Vaccines based on B.1.1.7 (alpha) are likely to provide the broadest protection against current variants.
- Published
- 2021
16. The molecular reach of antibodies crucially underpins their viral neutralisation capacity
- Author
-
Huhn, Anna, Nissley, Daniel, Wilson, Daniel B., Kutuzov, Mikhail A., Donat, Robert, Tan, Tiong Kit, Zhang, Ying, Barton, Michael I., Liu, Chang, Dejnirattisai, Wanwisa, Supasa, Piyada, Mongkolsapaya, Juthathip, Townsend, Alain, James, William, Screaton, Gavin, van der Merwe, P. Anton, Deane, Charlotte M., Isaacson, Samuel A., and Dushek, Omer
- Published
- 2025
- Full Text
- View/download PDF
17. Evidence of escape of SARS-CoV-2 variant B.1.351 from natural and vaccine-induced sera
- Author
-
Derrick W. Crook, Eleanor Barnes, Naomi Coombes, D. Zhou, Jingshan Ren, Duyvesteyn Hme., Bassam Hallis, Alexander J. Mentzer, Christina Dold, Sheila F Lumley, Cesar Lopez-Camacho, Aekkachai Tuekprakhon, Elizabeth E. Fry, Guido C. Paesen, Thomas S. Walter, Donal T. Skelly, Beibei Wang, Helen M. Ginn, David I. Stuart, Chang Liu, Sandra Belij-Rammerstorfer, Julian C. Knight, Susanna Dunachie, Juthathip Mongkolsapaya, S Bibi, Teresa Lambe, Amy Flaxman, Sarah C. Gilbert, P Supasa, Wanwisa Dejnirattisai, Paul Klenerman, M Bittaye, Miles W. Carroll, Tao Dong, Sue Charlton, R Levin, R Nutalai, Yuguang Zhao, William James, Gavin R. Screaton, Kevin R. Bewley, Elizabeth A. Clutterbuck, J Slon-Campos, and Andrew J. Pollard
- Subjects
Models, Molecular ,COVID-19 Vaccines ,medicine.drug_class ,Plasma protein binding ,Biology ,Monoclonal antibody ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,03 medical and health sciences ,0302 clinical medicine ,Neutralization Tests ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Receptor ,Vero Cells ,COVID-19 Serotherapy ,030304 developmental biology ,0303 health sciences ,Mutation ,Clinical Trials as Topic ,SARS-CoV-2 ,HEK 293 cells ,Immunization, Passive ,Antibodies, Monoclonal ,COVID-19 ,Virology ,HEK293 Cells ,biology.protein ,Vero cell ,Antibody ,030217 neurology & neurosurgery ,Protein Binding - Abstract
The race to produce vaccines against SARS-CoV-2 began when the first sequence was published, and this forms the basis for vaccines currently deployed globally. Independent lineages of SARS-CoV-2 have recently been reported: UK–B.1.1.7, South Africa–B.1.351 and Brazil–P.1. These variants have multiple changes in the immunodominant spike protein which facilitates viral cell entry via the Angiotensin converting enzyme-2 (ACE2) receptor. Mutations in the receptor recognition site on the spike are of great concern for their potential for immune escape. Here we describe a structure-function analysis of B.1.351 using a large cohort of convalescent and vaccinee serum samples. The receptor binding domain mutations provide tighter ACE2 binding and widespread escape from monoclonal antibody neutralization largely driven by E484K although K417N and N501Y act together against some important antibody classes. In a number of cases it would appear that convalescent and some vaccine serum offers limited protection against this variant., Structure-function analysis of the SARS-CoV-2 variant B.1.351 using serum samples from convalescent and vaccinated individuals reveals how mutations in the viral spike protein result in tighter binding to the receptor ACE2 and allow escape from monoclonal antibody neutralization.
- Published
- 2021
18. Reduced neutralization of SARS-CoV-2 B.1.1.7 variant by convalescent and vaccine sera
- Author
-
Aekkachai Tuekprakhon, Amy Flaxman, Sarah C. Gilbert, Mark A. Williams, Teresa Lambe, Guido C. Paesen, J Slon-Campos, Eleanor Barnes, William James, N Gent, Naomi Coombes, I Shaik, Sue Charlton, Andrew J. Pollard, Christina Dold, Sheila F Lumley, A Sienkiewicz, Bassam Hallis, Alexander J. Mentzer, D. Zhou, Elizabeth E. Fry, R Nutalai, Kerry J Godwin, Gavin R. Screaton, Cesar Lopez-Camacho, Kevin R. Bewley, Paul Klenerman, Elizabeth A. Clutterbuck, Miles W. Carroll, Wanwisa Dejnirattisai, S Bibi, Beibei Wang, Juthathip Mongkolsapaya, Susanna Dunachie, Sandra Belij-Rammerstorfer, Jingshan Ren, David R. Hall, R Levin, Thomas S. Walter, Donal T. Skelly, Natalie Baker, Helen M. Ginn, P Supasa, Tao Dong, Chang Liu, Neil G. Paterson, Yuguang Zhao, Julian C. Knight, Holly E. Humphries, M Bittaye, David I. Stuart, D W Crook, and Duyvesteyn Hme.
- Subjects
medicine.drug_class ,CHO Cells ,Biology ,Antibodies, Viral ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,Neutralization ,Virus ,Article ,Structure-Activity Relationship ,03 medical and health sciences ,Cricetulus ,0302 clinical medicine ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Pandemics ,Vero Cells ,030304 developmental biology ,Vaccines ,0303 health sciences ,SARS-CoV-2 ,Transmission (medicine) ,Antibodies, Monoclonal ,COVID-19 ,Antibodies, Neutralizing ,Research Highlight ,Virology ,Vaccination ,HEK293 Cells ,Viral replication ,Spike Glycoprotein, Coronavirus ,Vero cell ,biology.protein ,Infectious diseases ,Antibody ,030217 neurology & neurosurgery ,Protein Binding - Abstract
SARS-CoV-2 has caused over 2M deaths in little over a year. Vaccines are being deployed at scale, aiming to generate responses against the virus spike. The scale of the pandemic and error-prone virus replication is leading to the appearance of mutant viruses and potentially escape from antibody responses. Variant B.1.1.7, now dominant in the UK, with increased transmission, harbours 9 amino-acid changes in the spike, including N501Y in the ACE2 interacting-surface. We examine the ability of B.1.1.7 to evade antibody responses elicited by natural SARS-CoV-2 infection or vaccination. We map the impact of N501Y by structure/function analysis of a large panel of well-characterised monoclonal antibodies. B.1.1.7 is harder to neutralize than parental virus, compromising neutralization by some members of a major class of public antibodies through light chain contacts with residue 501. However, widespread escape from monoclonal antibodies or antibody responses generated by natural infection or vaccination was not observed., The SARS-CoV-2 B.1.1.7 variant is not neutralized as easily as the original form of the virus. Some public antibodies cannot neutralize B.1.1.7, due to altered light chain contacts with residue 501. However, B.1.1.7 does not show widespread escape from monoclonal antibodies, natural antibody responses, or vaccines.
- Published
- 2021
19. Native-like SARS-CoV-2 spike glycoprotein expressed by ChAdOx1 nCoV-19/AZD1222 vaccine
- Author
-
Mercede Lee, Joel D. Allen, Wanwisa Dejnirattisai, David Pulido, Francesca R. Donnellan, Yasunori Watanabe, Max Crispin, Peijun Zhang, Catherine M. Green, Marta Ulaszewska, Hannah Davies, Juthathip Mongkolsapaya, Teresa Lambe, Gavin R. Screaton, Sarah C. Gilbert, Krebs A-S., Himanshi Chawla, A Howe, Sandra Belij-Rammerstorfer, P Supasa, Susan J. Morris, Luiza Mendonça, and Elizabeth R. Allen
- Subjects
2019-20 coronavirus outbreak ,Glycosylation ,General Chemical Engineering ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,Viral antigen ,010402 general chemistry ,01 natural sciences ,Virus ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,QD1-999 ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,010405 organic chemistry ,General Chemistry ,Virology ,0104 chemical sciences ,3. Good health ,Chemistry ,chemistry ,biology.protein ,Antibody ,Glycoprotein ,030217 neurology & neurosurgery ,Research Article - Abstract
Vaccine development against the SARS-CoV-2 virus focuses on the principal target of the neutralizing immune response, the spike (S) glycoprotein. Adenovirus-vectored vaccines offer an effective platform for the delivery of viral antigen, but it is important for the generation of neutralizing antibodies that they produce appropriately processed and assembled viral antigen that mimics that observed on the SARS-CoV-2 virus. Here, we describe the structure, conformation, and glycosylation of the S protein derived from the adenovirus-vectored ChAdOx1 nCoV-19/AZD1222 vaccine. We demonstrate native-like post-translational processing and assembly, and reveal the expression of S proteins on the surface of cells adopting the trimeric prefusion conformation. The data presented here confirm the use of ChAdOx1 adenovirus vectors as a leading platform technology for SARS-CoV-2 vaccines., The structure, conformation, and glycosylation of the S protein derived from the adenovirus-vectored ChAdOx1 nCoV-19/AZD1222 vaccine are described. Also, native-like post-translational processing and assembly are demonstrated.
- Published
- 2021
20. Sustained T Cell Immunity, Protection and Boosting Using Extended Dosing Intervals of BNT162b2 mRNA Vaccine
- Author
-
Katie Jeffery, Alex G. Richter, Rebecca Brown, Susanna Dunachie, Adrian M Shields, Susan Hopkins, Sarah Foulkes, S Al-Taei, A R Nicols, T Malone, S A Johnson, Wanwisa Dejnirattisai, H Hornsby, J K Tyerman, Christina Dold, Eleanor Barnes, V J Hall, Donal T. Skelly, J A Kilby, G Sandhar, Shona C Moore, Anthony Brown, S Adele, Thushan de Silva, Adrienn Angyal, Daniel G. Wootton, T Tipton, P Supasa, Ali Amini, Alexandra Deeks, Andy Hargreaves, John Frater, R Whitham, Christopher P. Conlon, Rebecca Payne, L M Hering, T Altmann, Ayoub Saei, Juthathip Mongkolsapaya, L Stafford, Christopher J A Duncan, S Longet, Paul M. Matthews, Andrew J. Pollard, Sian E Faustini, J A Austin, Natalie Gillson, A Cross, N Meardon, Paul Klenerman, Elizabeth Phillips, L Turtle, S L Dobson, Sarah Rowland-Jones, Gavin R. Screaton, and Michael C. Carroll
- Subjects
medicine.medical_specialty ,education.field_of_study ,Research ethics ,business.industry ,Public health ,education ,Population ,Clinical trial ,Informed consent ,Family medicine ,Good clinical practice ,Health care ,medicine ,business ,Declaration of Helsinki - Abstract
Extension of the interval between vaccine doses for the BNT162b2 mRNA vaccine was introduced in the UK to accelerate population coverage with a single dose. In a study of 503 healthcare workers, we show that after priming following the first vaccine there is a marked decline in SARS-CoV-2 neutralizing antibody (NAb) levels, but, in contrast, a sustained T cell response to spike protein. This divergent immune profile was accompanied by robust protection from infection over this period from the circulating alpha (B.1.1.7) variant. Importantly, following the second vaccine dose, NAb levels were higher after the extended dosing interval (6-14 weeks) compared to the conventional 3-4 week regimen, accompanied by a clear enrichment of CD4+ T cells expressing IL2. These data on dynamic cellular and humoral responses indicate that extension of the dosing interval is an effective, immunogenic protocol and that antiviral T cell responses are a potential mechanism of protection.Trial Registration Details: PITCH is a sub-study of the SIREN study which is registered with ISRCTN, number ISRCTN11041050,Funding Information: This work was funded by the UK Department of Health and Social Care as part of the PITCH (Protective Immunity from T cells to Covid-19 in Health workers) Consortium, with contributions from UKRI/NIHR through the UK Coronavirus Immunology Consortium (UK-CIC), the Huo Family Foundation and The National Institute for Health Research (UKRIDHSC COVID-19 Rapid Response Rolling Call, Grant Reference Number COV19-RECPLAS).EB and PK are NIHR Senior Investigators and PK is funded by WT109965MA. SJD is funded by an NIHR Global Research Professorship (NIHR300791). TdS is funded by a Wellcome Trust Intermediate Clinical Fellowship (110058/Z/15/Z). RPP is funded by a Career Re-entry Fellowship (204721/Z/16/Z). CJAD is funded by a Wellcome Clinical Research Career Development Fellowship (211153/Z/18/Z). DS is supported by the NIHR Academic Clinical Lecturer programme in Oxford. LT is supported by the Wellcome Trust (grant number 205228/Z/16/Z) and the National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Emerging and Zoonotic Infections (NIHR200907) at University of Liverpool in partnership with Public Health England (PHE), in collaboration with Liverpool School of Tropical Medicine and the University of Oxford. DGW is supported by an NIHR Advanced Fellowship in Liverpool. LT and MC are supported by U.S. Food and Drug Administration Medical Countermeasures Initiative contract 75F40120C00085. Declaration of Interests: AJP is Chair of UK Dept. Health and Social Care’s (DHSC) Joint Committee on Vaccination & Immunisation (JCVI), but does not participate in policy decisions on COVID-19 vaccines. He is a member of the WHO’s SAGE. The views expressed in this article do not necessarily represent the views of DHSC, JCVI, or WHO. AJP is chief investigator on clinical trials of Oxford University’s COVID-19 vaccine funded by NIHR. Oxford University has entered a joint COVID-19 vaccine development partnership with AstraZeneca. Ethics Approval Statement: PITCH is a sub-study of the SIREN study which was approved by the Berkshire Research Ethics Committee, Health Research 250 Authority (IRAS ID 284460, REC reference 20/SC/0230), with PITCH recognised as a sub-study on 2 December 2020. SIREN is registered with ISRCTN (Trial ID:252 ISRCTN11041050). Some participants were recruited under aligned study protocols. In Birmingham participants were recruited under the Determining the immune response to SARS-CoV-2 infection in convalescent health care workers (COCO) study (IRAS ID: 282525). In Liverpool some participants were recruited under the “Human immune responses to acute virus infections” Study (16/NW/0170), approved by North West - Liverpool Central Research Ethics Committee on 8 March 2016, and amended on 14th September 2020 and 4th May 2021. In Oxford, participants were recruited under the GI Biobank Study 16/YH/0247, approved by the research ethics committee (REC) at Yorkshire & The Humber - Sheffield Research Ethics Committee on 29 July 2016, which has been amended for this purpose on 8 June 2020. In Sheffield, participants were recruited under the Observational Biobanking study STHObs (18/YH/0441), which was amended for this study on 10 September 2020. The study was conducted in compliance with all relevant ethical regulations for work with human participants, and according to the principles of the Declaration of Helsinki (2008) and the International Conference on Harmonization (ICH) Good Clinical Practice (GCP) guidelines. Written informed consent was obtained for all patients enrolled in the study.
- Published
- 2021
- Full Text
- View/download PDF
21. A haemagglutination test for rapid detection of antibodies to SARS-CoV-2
- Author
-
V Gallardo Sanchez, T Lockett, Rutger J. Ploeg, Denise O'Donnell, Shona C Moore, Julian C. Knight, Elizabeth A. Clutterbuck, G Scozzafava, Abigail Lamikanra, Sarah Hoosdally, S Bibi, Philippa C Matthews, Matt J. Neville, Huang K-Ya., Christina Dold, F Karpe, Christopher P. Conlon, A Allcock, Jose Vicente Martinez, Mark A. Ainsworth, John Frater, Donal T. Skelly, Juthathip Mongkolsapaya, A Sobrinodiaz, E Perez, Susanna Dunachie, Carolina V. Arancibia-Cárcamo, Richard Vipond, Jiangdong Huo, Marta S Oliveira, Openshaw Pjm., Sally Beer, Maria Fernandez Mendoza, Justine K. Rudkin, Etienne Joly, Derrick W. Crook, Teresa L Street, Abbie Bown, A Espinosa, Julie Xiao, Rolle Rahikainen, Turtle Lcw., J K Baillie, H P Tsang, Lisa Schimanski, Katie Jeffrey, Andrew J Kwok, P Supasa, Nicole Stoesser, H Thraves, Nimesh Gupta, D Georgiou, Pramila Rijal, Malcolm G Semple, Paul Klenerman, Eleanor Barnes, Alain Townsend, Gavin R. Screaton, L Stafford, David J. Roberts, Wanwisa Dejnirattisai, Tiong Kit Tan, and Alexander J. Mentzer
- Subjects
0301 basic medicine ,Hemagglutination ,Epidemiology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Point-of-care testing ,Science ,Point-of-Care Systems ,General Physics and Astronomy ,Enzyme-Linked Immunosorbent Assay ,Red cell agglutination ,Antibodies, Viral ,Polymerase Chain Reaction ,Sensitivity and Specificity ,General Biochemistry, Genetics and Molecular Biology ,Epitope ,Article ,Antibodies ,Serology ,03 medical and health sciences ,0302 clinical medicine ,COVID-19 Testing ,Agglutination Tests ,Medicine ,Humans ,Seroconversion ,Point of care ,Multidisciplinary ,biology ,Red Cell ,business.industry ,SARS-CoV-2 ,fungi ,Antibodies, Monoclonal ,COVID-19 ,Diagnostic markers ,General Chemistry ,Hemagglutination Tests ,Virology ,Test (assessment) ,030104 developmental biology ,Spike Glycoprotein, Coronavirus ,biology.protein ,Antibody ,business ,030217 neurology & neurosurgery - Abstract
Serological detection of antibodies to SARS-CoV-2 is essential for establishing rates of seroconversion in populations, and for seeking evidence for a level of antibody that may be protective against COVID-19 disease. Several high-performance commercial tests have been described, but these require centralised laboratory facilities that are comparatively expensive, and therefore not available universally. Red cell agglutination tests do not require special equipment, are read by eye, have short development times, low cost and can be applied at the Point of Care. Here we describe a quantitative Haemagglutination test (HAT) for the detection of antibodies to the receptor binding domain of the SARS-CoV-2 spike protein. The HAT has a sensitivity of 90% and specificity of 99% for detection of antibodies after a PCR diagnosed infection. We will supply aliquots of the test reagent sufficient for ten thousand test wells free of charge to qualified research groups anywhere in the world., Serological detection of antibodies against SARS-CoV-2 can help establish rates of seroconversion. Here the authors develop a red cell agglutination test to detect antibodies against the receptor binding domain for distribution free of charge to qualified research groups.
- Published
- 2020
- Full Text
- View/download PDF
22. Structural basis for the neutralization of SARS-CoV-2 by an antibody from a convalescent patient
- Author
-
Pramila Rijal, Loic Carrique, Neil G. Paterson, Elizabeth E. Fry, Chen C-P., Huang K-Ya., Alain Townsend, Gavin R. Screaton, Reinis R. Ruza, Shah Pnm., D. Zhou, Karen R. Buttigieg, Miles W. Carroll, William James, Raymond J. Owens, J. Huo, James H. Naismith, Jingshan Ren, Huang Y-C., Kerry J Godwin, Lin T-Y., Shih S-R., Chen T-H., Javier Gilbert-Jaramillo, Huang C-G., Cheng S-H., Julia A. Tree, Tomas Malinauskas, David I. Stuart, Che Ma, R F Donat, Julika Radecke, Tiong Kit Tan, Michael L. Knight, P Supasa, Cheng C-Y., Duyvesteyn Hme., Juthathip Mongkolsapaya, Lin Y-C., and Yuguang Zhao
- Subjects
Adult ,Male ,Coronavirus disease 2019 (COVID-19) ,Protein Conformation ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Protein domain ,Trimer ,Cross Reactions ,Peptidyl-Dipeptidase A ,Antibodies, Viral ,Crystallography, X-Ray ,Receptor binding site ,Neutralization ,Epitope ,Betacoronavirus ,Epitopes ,Immunoglobulin Fab Fragments ,03 medical and health sciences ,0302 clinical medicine ,Protein structure ,Protein Domains ,Structural Biology ,Chlorocebus aethiops ,Animals ,Humans ,Binding site ,Pandemics ,Vero Cells ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Binding Sites ,biology ,SARS-CoV-2 ,Cryoelectron Microscopy ,COVID-19 ,Antibodies, Neutralizing ,Virology ,chemistry ,Spike Glycoprotein, Coronavirus ,biology.protein ,Angiotensin-Converting Enzyme 2 ,Antibody ,Coronavirus Infections ,Glycoprotein ,030217 neurology & neurosurgery - Abstract
The COVID-19 pandemic has had unprecedented health and economic impact, but currently there are no approved therapies. We have isolated an antibody, EY6A, from a late-stage COVID-19 patient and show it neutralises SARS-CoV-2 and cross-reacts with SARS-CoV-1. EY6A Fab binds tightly (KD of 2 nM) the receptor binding domain (RBD) of the viral Spike glycoprotein and a 2.6Å crystal structure of an RBD/EY6A Fab complex identifies the highly conserved epitope, away from the ACE2 receptor binding site. Residues of this epitope are key to stabilising the pre-fusion Spike. Cryo-EM analyses of the pre-fusion Spike incubated with EY6A Fab reveal a complex of the intact trimer with three Fabs bound and two further multimeric forms comprising destabilized Spike attached to Fab. EY6A binds what is probably a major neutralising epitope, making it a candidate therapeutic for COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
23. Broad and strong memory CD4 (+) and CD8 (+) T cells induced by SARS-CoV-2 in UK convalescent COVID-19 patients
- Author
-
Tao Dong, A Ansari, Persephone Borrow, Cesar Lopez-Camacho, Ker D-S., Openshaw Pjm., P Goulder, Juthathip Mongkolsapaya, Peter Simmonds, Gavin R. Screaton, Giorgio Napolitani, R Levin, D Dong, Eleanor Barnes, Peijun Zhang, Graham S. Ogg, Kenneth Baillie, P Supasa, Susanna Dunachie, G Paeson, Gu Liu, Georgina Kerr, Jeremy Ratcliff, J Slon-Campos, Wanwisa Dejnirattisai, Zixi Yin, T Lockett, Lance Turtle, Dannielle Wellington, X Yao, Richard J. Cornall, U S Rajapaksa, J W Fry, Julian C. Knight, Christopher P. Conlon, Jonathan M. Grimes, Christina Dold, Paul Sopp, Mariolina Salio, Andrew J. McMichael, F Antson, John Frater, Yanchun Peng, Malcolm G Semple, Paul Klenerman, Wayne Paes, T I de Silva, Benedikt M. Kessler, David I. Stuart, Oliver W. Bayfield, Krishanthi Subramaniam, Chang Liu, Shona C Moore, Yuguang Zhao, N F Schwabe, Alexander J. Mentzer, Chen Y-L., Paul Thomson, T Rostron, and Hawkins Dedp.
- Subjects
Innate immune system ,T cell ,Biology ,Virus ,Epitope ,Article ,medicine.anatomical_structure ,Immunology ,medicine ,biology.protein ,Cytotoxic T cell ,Antibody ,Memory T cell ,CD8 - Abstract
COVID-19 is an ongoing global crisis in which the development of effective vaccines and therapeutics will depend critically on understanding the natural immunity to the virus, including the role of SARS-CoV-2-specific T cells. We have conducted a study of 42 patients following recovery from COVID-19, including 28 mild and 14 severe cases, comparing their T cell responses to those of 16 control donors. We assessed the immune memory of T cell responses using IFNγ based assays with overlapping peptides spanning SARS-CoV-2 apart from ORF1. We found the breadth, magnitude and frequency of memory T cell responses from COVID-19 were significantly higher in severe compared to mild COVID-19 cases, and this effect was most marked in response to spike, membrane, and ORF3a proteins. Total and spike-specific T cell responses correlated with the anti-Spike, anti-Receptor Binding Domain (RBD) as well as anti-Nucleoprotein (NP) endpoint antibody titre (p+and/or CD8+epitopes, which strikingly included six immunodominant epitope clusters targeted by T cells in many donors, including 3 clusters in spike (recognised by 29%, 24%, 18% donors), two in the membrane protein (M, 32%, 47%) and one in the nucleoprotein (Np, 35%). CD8+ responses were further defined for their HLA restriction, including B*4001-restricted T cells showing central memory and effector memory phenotype. In mild cases, higher frequencies of multi-cytokine producing M- and NP-specific CD8+T cells than spike-specific CD8+T cells were observed. They furthermore showed a higher ratio of SARS-CoV-2-specific CD8+to CD4+T cell responses. Immunodominant epitope clusters and peptides containing T cell epitopes identified in this study will provide critical tools to study the role of virus-specific T cells in control and resolution of SARS-CoV-2 infections. The identification of T cell specificity and functionality associated with milder disease, highlights the potential importance of including non-spike proteins within future COVID-19 vaccine design.
- Published
- 2020
24. Neutralization of SARS-CoV-2 by destruction of the prefusion Spike
- Author
-
Raymond J. Owens, Alain Townsend, Naomi Coombes, D. Zhou, Neil G. Paterson, Pramila Rijal, Reinis R. Ruza, Kevin R. Bewley, Loic Carrique, Julika Radecke, David I. Stuart, Tiong Kit Tan, Tomas Malinauskas, Juthathip Mongkolsapaya, Elizabeth E. Fry, Yuguang Zhao, Jiandong Huo, Helen M. E. Duyvesteyn, Miles W. Carroll, P Supasa, Gavin R. Screaton, Helen M. Ginn, Jingshan Ren, and Pranav N.M. Shah
- Subjects
0303 health sciences ,050208 finance ,biology ,Chemistry ,medicine.drug_class ,05 social sciences ,Lipid bilayer fusion ,medicine.disease_cause ,Monoclonal antibody ,Epitope ,Neutralization ,3. Good health ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,0502 economics and business ,medicine ,biology.protein ,030212 general & internal medicine ,050207 economics ,Antibody ,Receptor ,030304 developmental biology ,Coronavirus - Abstract
SummaryThere are as yet no licenced therapeutics for the COVID-19 pandemic. The causal coronavirus (SARS-CoV-2) binds host cells via a trimeric Spike whose receptor binding domain (RBD) recognizes angiotensin-converting enzyme 2 (ACE2), initiating conformational changes that drive membrane fusion. We find that monoclonal antibody CR3022 binds the RBD tightly, neutralising SARS-CoV-2 and report the crystal structure at 2.4 Å of the Fab/RBD complex. Some crystals are suitable for screening for entry-blocking inhibitors. The highly conserved, structure-stabilising, CR3022 epitope is inaccessible in the prefusion Spike, suggesting that CR3022 binding would facilitate conversion to the fusion-incompetent post-fusion state. Cryo-EM analysis confirms that incubation of Spike with CR3022 Fab leads to destruction of the prefusion trimer. Presentation of this cryptic epitope in an RBD-based vaccine might advantageously focus immune responses. Binders at this epitope may be useful therapeutically, possibly in synergy with an antibody blocking receptor attachment.HighlightsCR3022 neutralises SARS-CoV-2Neutralisation is by destroying the prefusion SPIKE conformationThis antibody may have therapeutic potential alone or with one blocking receptor attachment
- Published
- 2020
- Full Text
- View/download PDF
25. Neutralization of SARS-CoV-2 by Destruction of the Prefusion Spike
- Author
-
Reinis R. Ruza, Neil G. Paterson, Elizabeth E. Fry, Yuguang Zhao, Miles W. Carroll, Helen M. E. Duyvesteyn, Alain Townsend, Kevin R. Bewley, Jingshan Ren, Julia A. Tree, Pramila Rijal, Loic Carrique, Helen M. Ginn, Raymond J. Owens, Tomas Malinauskas, David I. Stuart, Gavin R. Screaton, P Supasa, Jiandong Huo, Juthathip Mongkolsapaya, Naomi Coombes, D. Zhou, Pranav N.M. Shah, Julika Radecke, and Tiong Kit Tan
- Subjects
Models, Molecular ,Antigen-Antibody Complex ,Antibodies, Viral ,Crystallography, X-Ray ,medicine.disease_cause ,Neutralization ,Epitope ,CR3022 ,0302 clinical medicine ,antibody ,Spike (database) ,Peptide sequence ,health care economics and organizations ,Confusion ,Coronavirus ,epitope ,0303 health sciences ,receptor binding domain ,Antibodies, Monoclonal ,Cell biology ,3. Good health ,Ectodomain ,Research centre ,Spike Glycoprotein, Coronavirus ,Receptors, Virus ,Angiotensin-Converting Enzyme 2 ,Antibody ,medicine.symptom ,Coronavirus Infections ,Allosteric Site ,COVID-19 Vaccines ,Coronavirus disease 2019 (COVID-19) ,medicine.drug_class ,Viral protein ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Allosteric regulation ,cryo-electron microscopy ,Peptidyl-Dipeptidase A ,Biology ,Monoclonal antibody ,Microbiology ,Article ,Betacoronavirus ,03 medical and health sciences ,Neutralization Tests ,Virology ,medicine ,Humans ,Amino Acid Sequence ,Pandemics ,X-ray crystallography ,030304 developmental biology ,Host Microbial Interactions ,Competing interests ,SARS-CoV-2 ,Host (biology) ,Cryoelectron Microscopy ,COVID-19 ,Lipid bilayer fusion ,Correction ,Viral Vaccines ,Regret ,spike ,neutralization ,Virus Internalization ,Antibodies, Neutralizing ,COVID-19 Drug Treatment ,therapeutic ,biology.protein ,Parasitology ,030217 neurology & neurosurgery - Abstract
Summary There are as yet no licensed therapeutics for the COVID-19 pandemic. The causal coronavirus (SARS-CoV-2) binds host cells via a trimeric spike whose receptor binding domain (RBD) recognizes angiotensin-converting enzyme 2, initiating conformational changes that drive membrane fusion. We find that the monoclonal antibody CR3022 binds the RBD tightly, neutralizing SARS-CoV-2, and report the crystal structure at 2.4 Å of the Fab/RBD complex. Some crystals are suitable for screening for entry-blocking inhibitors. The highly conserved, structure-stabilizing CR3022 epitope is inaccessible in the prefusion spike, suggesting that CR3022 binding facilitates conversion to the fusion-incompetent post-fusion state. Cryogenic electron microscopy (cryo-EM) analysis confirms that incubation of spike with CR3022 Fab leads to destruction of the prefusion trimer. Presentation of this cryptic epitope in an RBD-based vaccine might advantageously focus immune responses. Binders at this epitope could be useful therapeutically, possibly in synergy with an antibody that blocks receptor attachment., Graphical Abstract, Highlights • CR3022 binds the RBD of SARS-CoV-2 and shows strong neutralization • Neutralization is by destroying the prefusion spike conformation • CR3022 binds a highly conserved epitope that is inaccessible in prefusion spike protein • CR3022 could have therapeutic potential alone or in synergy with a receptor blocker, Huo et al. find that the antibody CR3022 binds tightly to the receptor binding domain of the SARS-CoV-2 spike at a site different to that used by the receptor. CR3022 effectively neutralizes the virus, and cryo-EM reveals that it disrupts the spike. Such antibodies could have potential as COVID-19 therapeutics.
- Published
- 2020
- Full Text
- View/download PDF
26. Antibody testing for COVID-19: A report from the National COVID Scientific Advisory Panel
- Author
-
Tessa Prince, Derrick W. Crook, Gavin R. Screaton, A Espinosa, P Supasa, Cesar Lopez-Camacho, Juthathip Mongkolsapaya, Nicholas A. Watkins, H Thraves, Philippa C Matthews, D Georgiou, Pat Tsang, Beibei Wang, Mark A. Ainsworth, Veronica Sanchez, Malcolm G Semple, Marta S Oliveira, Wanwisa Dejnirattisai, Anne-Sophie Walker, Rutger J. Ploeg, J Milton, Tomas Surik, C Knowles, Andrew J. Pollard, Julian C. Knight, Kathryn Auckland, J K Baillie, Andrew J Kwok, Paul Klenerman, Alexander J. Mentzer, David I. Stuart, Fiona Pereira, Donal T. Skelly, Senthil Chinnakannan, H McGivern, Eleanor Barnes, John I. Bell, S Bibi, Rekha Anand, T Peto, Justine K. Rudkin, U Leuschner, C Washington, Lance Turtle, E Perez, T Berry, Monique Andersson, Sally Beer, Emily R. Adams, A Sobrinodiaz, T I de Silva, José William Martínez, D F Kelly, Chang Liu, J Whitehouse, Shona C Moore, K Jefferey, R Levin, J Slon-Campos, Julie Staves, A Hunter, H Farmer, M Fernandez Mendoza, Richard J. Cornall, Elizabeth A. Clutterbuck, Devender Roberts, David W Eyre, E. N. Smith, Sarah Hoosdally, Kate E. Dingle, Christina Dold, and Miles W. Carroll
- Subjects
0301 basic medicine ,medicine.medical_specialty ,IgM ,Coronavirus disease 2019 (COVID-19) ,IgG ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Population ,serology ,Medicine (miscellaneous) ,Gastroenterology ,General Biochemistry, Genetics and Molecular Biology ,Serology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,antibodies ,Medicine ,immunoassay ,030212 general & internal medicine ,education ,education.field_of_study ,biology ,medicine.diagnostic_test ,SARS-CoV-2 ,business.industry ,COVID-19 ,virus diseases ,Articles ,biochemical phenomena, metabolism, and nutrition ,030104 developmental biology ,lateral flow ,exposure ,Immunoassay ,biology.protein ,ELISA ,epidemiology ,Antibody ,business ,Research Article ,Lateral flow immunoassay ,Antibody detection - Abstract
Background: The COVID-19 pandemic caused >1 million infections during January-March 2020. There is an urgent need for reliable antibody detection approaches to support diagnosis, vaccine development, safe release of individuals from quarantine, and population lock-down exit strategies. We set out to evaluate the performance of ELISA and lateral flow immunoassay (LFIA) devices. Methods: We tested plasma for COVID (severe acute respiratory syndrome coronavirus 2; SARS-CoV-2) IgM and IgG antibodies by ELISA and using nine different LFIA devices. We used a panel of plasma samples from individuals who have had confirmed COVID infection based on a PCR result (n=40), and pre-pandemic negative control samples banked in the UK prior to December-2019 (n=142). Results: ELISA detected IgM or IgG in 34/40 individuals with a confirmed history of COVID infection (sensitivity 85%, 95%CI 70-94%), vs. 0/50 pre-pandemic controls (specificity 100% [95%CI 93-100%]). IgG levels were detected in 31/31 COVID-positive individuals tested ≥10 days after symptom onset (sensitivity 100%, 95%CI 89-100%). IgG titres rose during the 3 weeks post symptom onset and began to fall by 8 weeks, but remained above the detection threshold. Point estimates for the sensitivity of LFIA devices ranged from 55-70% versus RT-PCR and 65-85% versus ELISA, with specificity 95-100% and 93-100% respectively. Within the limits of the study size, the performance of most LFIA devices was similar. Conclusions: Currently available commercial LFIA devices do not perform sufficiently well for individual patient applications. However, ELISA can be calibrated to be specific for detecting and quantifying SARS-CoV-2 IgM and IgG and is highly sensitive for IgG from 10 days following first symptoms.
- Published
- 2020
- Full Text
- View/download PDF
27. Heterologous versus homologous COVID-19 booster vaccination in previous recipients of two doses of CoronaVac COVID-19 vaccine in Brazil (RHH-001): a phase 4, non-inferiority, single blind, randomised study
- Author
-
Costa Clemens, Sue Ann, Weckx, Lily, Clemens, Ralf, Almeida Mendes, Ana Verena, Ramos Souza, Alessandra, Silveira, Mariana B V, da Guarda, Suzete Nascimento Farias, de Nobrega, Maristela Miyamoto, de Moraes Pinto, Maria Isabel, Gonzalez, Isabela G S, Salvador, Natalia, Franco, Marilia Miranda, de Avila Mendonça, Renata Navis, Queiroz Oliveira, Isabelle Silva, de Freitas Souza, Bruno Solano, Fraga, Mayara, Aley, Parvinder, Bibi, Sagida, Cantrell, Liberty, Dejnirattisai, Wanwisa, Liu, Xinxue, Mongkolsapaya, Juthathip, Supasa, Piyada, Screaton, Gavin R, Lambe, Teresa, Voysey, Merryn, Pollard, Andrew J, Bittaye, Mustapha, Woods, Danielle, Davies, Sophie, Smith, Holly, Ulaszewska, Marta, Sanders, Helen, Mabette, Reece, Vernon, Sophie, Valliji, Zara, Mead, Gracie, Tejpal, Chitra, Park, Juyeon, Beveridge, Amy, Eldawi, Ahmed, Felle, Sally, Fraga, Mayara, Muniz Martins, Thaiane, Martins Medrado, Claudia Loureiro, and de Arruda Cordeiro Matos, Laiana Januse
- Abstract
The inactivated whole-virion SARS-CoV-2 vaccine (CoronaVac, Sinovac) has been widely used in a two-dose schedule. We assessed whether a third dose of the homologous or a different vaccine could boost immune responses.
- Published
- 2022
- Full Text
- View/download PDF
28. A structure-function analysis shows SARS-CoV-2 BA.2.86 balances antibody escape and ACE2 affinity
- Author
-
Liu, Chang, Zhou, Daming, Dijokaite-Guraliuc, Aiste, Supasa, Piyada, Duyvesteyn, Helen M.E., Ginn, Helen M., Selvaraj, Muneeswaran, Mentzer, Alexander J., Das, Raksha, de Silva, Thushan I., Ritter, Thomas G., Plowright, Megan, Newman, Thomas A.H., Stafford, Lizzie, Kronsteiner, Barbara, Temperton, Nigel, Lui, Yuan, Fellermeyer, Martin, Goulder, Philip, Klenerman, Paul, Dunachie, Susanna J., Barton, Michael I., Kutuzov, Mikhail A., Dushek, Omer, Fry, Elizabeth E., Mongkolsapaya, Juthathip, Ren, Jingshan, Stuart, David I., and Screaton, Gavin R.
- Abstract
BA.2.86, a recently described sublineage of SARS-CoV-2 Omicron, contains many mutations in the spike gene. It appears to have originated from BA.2 and is distinct from the XBB variants responsible for many infections in 2023. The global spread and plethora of mutations in BA.2.86 has caused concern that it may possess greater immune-evasive potential, leading to a new wave of infection. Here, we examine the ability of BA.2.86 to evade the antibody response to infection using a panel of vaccinated or naturally infected sera and find that it shows marginally less immune evasion than XBB.1.5. We locate BA.2.86 in the antigenic landscape of recent variants and look at its ability to escape panels of potent monoclonal antibodies generated against contemporary SARS-CoV-2 infections. We demonstrate, and provide a structural explanation for, increased affinity of BA.2.86 to ACE2, which may increase transmissibility.
- Published
- 2024
- Full Text
- View/download PDF
29. Comparative analysis of SARS-CoV-2 neutralization titers reveals consistency between human and animal model serum and across assays
- Author
-
Mühlemann, Barbara, Wilks, Samuel H., Baracco, Lauren, Bekliz, Meriem, Carreño, Juan Manuel, Corman, Victor M., Davis-Gardner, Meredith E., Dejnirattisai, Wanwisa, Diamond, Michael S., Douek, Daniel C., Drosten, Christian, Eckerle, Isabella, Edara, Venkata-Viswanadh, Ellis, Madison, Fouchier, Ron A. M., Frieman, Matthew, Godbole, Sucheta, Haagmans, Bart, Halfmann, Peter J., Henry, Amy R., Jones, Terry C., Katzelnick, Leah C., Kawaoka, Yoshihiro, Kimpel, Janine, Krammer, Florian, Lai, Lilin, Liu, Chang, Lusvarghi, Sabrina, Meyer, Benjamin, Mongkolsapaya, Juthathip, Montefiori, David C., Mykytyn, Anna, Netzl, Antonia, Pollett, Simon, Rössler, Annika, Screaton, Gavin R., Shen, Xiaoying, Sigal, Alex, Simon, Viviana, Subramanian, Rahul, Supasa, Piyada, Suthar, Mehul S., Türeli, Sina, Wang, Wei, Weiss, Carol D., and Smith, Derek J.
- Abstract
The evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires ongoing monitoring to judge the ability of newly arising variants to escape the immune response. A surveillance system necessitates an understanding of differences in neutralization titers measured in different assays and using human and animal serum samples. We compared 18 datasets generated using human, hamster, and mouse serum and six different neutralization assays. Datasets using animal model serum samples showed higher titer magnitudes than datasets using human serum samples in this comparison. Fold change in neutralization of variants compared to ancestral SARS-CoV-2, immunodominance patterns, and antigenic maps were similar among serum samples and assays. Most assays yielded consistent results, except for differences in fold change in cytopathic effect assays. Hamster serum samples were a consistent surrogate for human first-infection serum samples. These results inform the transition of surveillance of SARS-CoV-2 antigenic variation from dependence on human first-infection serum samples to the utilization of serum samples from animal models.
- Published
- 2024
- Full Text
- View/download PDF
30. Broad and strong memory CD4+and CD8+T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19
- Author
-
Peng, Yanchun, Mentzer, Alexander J., Liu, Guihai, Yao, Xuan, Yin, Zixi, Dong, Danning, Dejnirattisai, Wanwisa, Rostron, Timothy, Supasa, Piyada, Liu, Chang, López-Camacho, César, Slon-Campos, Jose, Zhao, Yuguang, Stuart, David I., Paesen, Guido C., Grimes, Jonathan M., Antson, Alfred A., Bayfield, Oliver W., Hawkins, Dorothy E. D. P., Ker, De-Sheng, Wang, Beibei, Turtle, Lance, Subramaniam, Krishanthi, Thomson, Paul, Zhang, Ping, Dold, Christina, Ratcliff, Jeremy, Simmonds, Peter, de Silva, Thushan, Sopp, Paul, Wellington, Dannielle, Rajapaksa, Ushani, Chen, Yi-Ling, Salio, Mariolina, Napolitani, Giorgio, Paes, Wayne, Borrow, Persephone, Kessler, Benedikt M., Fry, Jeremy W., Schwabe, Nikolai F., Semple, Malcolm G., Baillie, J. Kenneth, Moore, Shona C., Openshaw, Peter J. M., Ansari, M. Azim, Dunachie, Susanna, Barnes, Eleanor, Frater, John, Kerr, Georgina, Goulder, Philip, Lockett, Teresa, Levin, Robert, Zhang, Yonghong, Jing, Ronghua, Ho, Ling-Pei, Cornall, Richard J., Conlon, Christopher P., Klenerman, Paul, Screaton, Gavin R., Mongkolsapaya, Juthathip, McMichael, Andrew, Knight, Julian C., Ogg, Graham, and Dong, Tao
- Abstract
The development of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines and therapeutics will depend on understanding viral immunity. We studied T cell memory in 42 patients following recovery from COVID-19 (28 with mild disease and 14 with severe disease) and 16 unexposed donors, using interferon-γ-based assays with peptides spanning SARS-CoV-2 except ORF1. The breadth and magnitude of T cell responses were significantly higher in severe as compared with mild cases. Total and spike-specific T cell responses correlated with spike-specific antibody responses. We identified 41 peptides containing CD4+and/or CD8+epitopes, including six immunodominant regions. Six optimized CD8+epitopes were defined, with peptide–MHC pentamer-positive cells displaying the central and effector memory phenotype. In mild cases, higher proportions of SARS-CoV-2-specific CD8+T cells were observed. The identification of T cell responses associated with milder disease will support an understanding of protective immunity and highlights the potential of including non-spike proteins within future COVID-19 vaccine design.
- Published
- 2020
- Full Text
- View/download PDF
31. Cooperativity and induced oligomerization control the interaction of SARS-CoV-2 with its cellular receptor and patient-derived antibodies
- Author
-
Asor, Roi, Olerinyova, Anna, Burnap, Sean A., Kushwah, Manish S., Hensen, Mario, Vasiljevic, Snežana, Chang, Liu, Dejnirattisa, Wanwisa, Supasa, Piyada, Stuart, David I., Screaton, Gavin R., Zitzmann, Nicole, Benesch, Justin L.P., Struwe, Weston B., and Kukura, Philipp
- Published
- 2024
- Full Text
- View/download PDF
32. Reduced neutralisation of SARS-CoV-2 omicron B.1.1.529 variant by post-immunisation serum
- Author
-
Dejnirattisai, Wanwisa, Shaw, Robert H, Supasa, Piyada, Liu, Chang, Stuart, Arabella SV, Pollard, Andrew J, Liu, Xinxue, Lambe, Teresa, Crook, Derrick, Stuart, Dave I, Mongkolsapaya, Juthathip, Nguyen-Van-Tam, Jonathan S, Snape, Matthew D, and Screaton, Gavin R
- Published
- 2022
- Full Text
- View/download PDF
33. Sustainable energy and CO2 reduction policy in Thailand: An input–output approach from production- and consumption-based perspectives.
- Author
-
Supasa, Tharinya, Hsiau, Shu-San, Lin, Shih-Mo, Wongsapai, Wongkot, Chang, Kuei-Feng, and Wu, Jiunn-Chi
- Subjects
RENEWABLE energy sources ,CARBON dioxide ,EMISSIONS (Air pollution) ,CLIMATE change mitigation ,GROSS domestic product - Abstract
Energy shortages and CO 2 emissions reductions are critical contemporary challenges for Thailand. A consumption-based analysis provides crucial information that enables policymakers to more comprehensively understand the hidden contributors of energy demand and CO 2 in the economy. The other manufacturing, construction and food and beverage sectors were amongst the five largest contributors to energy use and emissions in both 2000 and 2010, based on a consumption perspective. However, these sectors have been neglected by energy conservation and climate change mitigation policies in Thailand because they were the least energy-intensive sectors per government energy reports from 1995 to 2015. The CO 2 emissions burden from exports was almost 50% of Thailand's national CO 2 inventory in 2000 and 2010. The embodied CO 2 emissions results revealed that Thailand could reduce its emissions inventory by 12% and 13% if embodied imports replaced exports in 2000 and 2010, respectively. Furthermore, the leading gross domestic product-generating industries in Thailand are seriously vulnerable to natural gas and crude oil shortages despite some sectors using them in small proportions in their production processes. Energy and emissions policies should better reflect consumption characteristics to increase the potential of energy-saving interventions and CO 2 mitigation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
34. Evaluation of T cell responses to naturally processed variant SARS-CoV-2 spike antigens in individuals following infection or vaccination
- Author
-
Yin, Zixi, Chen, Ji-Li, Lu, Yongxu, Wang, Beibei, Godfrey, Leila, Mentzer, Alexander J., Yao, Xuan, Liu, Guihai, Wellington, Dannielle, Zhao, Yiqi, Wing, Peter A.C., Dejnirattisa, Wanwisa, Supasa, Piyada, Liu, Chang, Hublitz, Philip, Beveridge, Ryan, Waugh, Craig, Clark, Sally-Ann, Clark, Kevin, Sopp, Paul, Rostron, Timothy, Mongkolsapaya, Juthathip, Screaton, Gavin R., Ogg, Graham, Ewer, Katie, Pollard, Andrew J., Gilbert, Sarah, Knight, Julian C., Lambe, Teresa, Smith, Geoffrey L., Dong, Tao, and Peng, Yanchun
- Abstract
Most existing studies characterizing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cell responses are peptide based. This does not allow evaluation of whether tested peptides are processed and presented canonically. In this study, we use recombinant vaccinia virus (rVACV)-mediated expression of SARS-CoV-2 spike protein and SARS-CoV-2 infection of angiotensin-converting enzyme (ACE)-2-transduced B cell lines to evaluate overall T cell responses in a small cohort of recovered COVID-19 patients and uninfected donors vaccinated with ChAdOx1 nCoV-19. We show that rVACV expression of SARS-CoV-2 antigen can be used as an alternative to SARS-CoV-2 infection to evaluate T cell responses to naturally processed spike antigens. In addition, the rVACV system can be used to evaluate the cross-reactivity of memory T cells to variants of concern (VOCs) and to identify epitope escape mutants. Finally, our data show that both natural infection and vaccination could induce multi-functional T cell responses with overall T cell responses remaining despite the identification of escape mutations.
- Published
- 2023
- Full Text
- View/download PDF
35. Rapid escape of new SARS-CoV-2 Omicron variants from BA.2-directed antibody responses
- Author
-
Dijokaite-Guraliuc, Aiste, Das, Raksha, Zhou, Daming, Ginn, Helen M., Liu, Chang, Duyvesteyn, Helen M.E., Huo, Jiandong, Nutalai, Rungtiwa, Supasa, Piyada, Selvaraj, Muneeswaran, de Silva, Thushan I., Plowright, Megan, Newman, Thomas A.H., Hornsby, Hailey, Mentzer, Alexander J., Skelly, Donal, Ritter, Thomas G., Temperton, Nigel, Klenerman, Paul, Barnes, Eleanor, Dunachie, Susanna J., Conlon, Christopher, Deeks, Alexandra, Frater, John, Gardiner, Siobhan, Jämsén, Anni, Jeffery, Katie, Malone, Tom, Phillips, Eloise, Kronsteiner-Dobramysl, Barbara, Abraham, Priyanka, Bibi, Sagida, Lambe, Teresa, Longet, Stephanie, Tipton, Tom, Carrol, Miles, Stafford, Lizzie, Roemer, Cornelius, Peacock, Thomas P., Paterson, Neil G., Williams, Mark A., Hall, David R., Fry, Elizabeth E., Mongkolsapaya, Juthathip, Ren, Jingshan, Stuart, David I., and Screaton, Gavin R.
- Abstract
In November 2021, Omicron BA.1, containing a raft of new spike mutations, emerged and quickly spread globally. Intense selection pressure to escape the antibody response produced by vaccines or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection then led to a rapid succession of Omicron sub-lineages with waves of BA.2 and then BA.4/5 infection. Recently, many variants have emerged such as BQ.1 and XBB, which carry up to 8 additional receptor-binding domain (RBD) amino acid substitutions compared with BA.2. We describe a panel of 25 potent monoclonal antibodies (mAbs) generated from vaccinees suffering BA.2 breakthrough infections. Epitope mapping shows potent mAb binding shifting to 3 clusters, 2 corresponding to early-pandemic binding hotspots. The RBD mutations in recent variants map close to these binding sites and knock out or severely knock down neutralization activity of all but 1 potent mAb. This recent mAb escape corresponds with large falls in neutralization titer of vaccine or BA.1, BA.2, or BA.4/5 immune serum.
- Published
- 2023
- Full Text
- View/download PDF
36. Evolution of long-term vaccine induced and hybrid immunity in healthcare workers after different COVID19 vaccine regimens: a longitudinal cohort study
- Author
-
Moore, Shona C., Kronsteiner, Barbara, Longet, Stephanie, Adele, Sandra, Deeks, Alexandra S., Liu, Chang, Dejnirattisai, Wanwisa, Reyes, Laura Silva, Meardon, Naomi, Faustini, Sian, Al-Taei, Saly, Tipton, Tom, Hering, Luisa M., Angyal, Adrienn, Brown, Rebecca, Nicols, Alexander R., Dobson, Susan L., Supasa, Piyada, Tuekprakhon, Aekkachai, Cross, Andrew, Tyerman, Jessica K., Hornsby, Hailey, Grouneva, Irina, Plowright, Megan, Zhang, Peijun, Newman, Thomas A.H., Nell, Jeremy M., Abraham, Priyanka, Ali, Mohammad, Malone, Tom, Neale, Isabel, Phillips, Eloise, Wilson, Joseph D., Murray, Sam M., Zewdie, Martha, Shields, Adrian, Horner, Emily C., Booth, Lucy H., Stafford, Lizzie, Bibi, Sagida, Wootton, Daniel G., Mentzer, Alexander J., Conlon, Christopher P., Jeffery, Katie, Matthews, Philippa C., Pollard, Andrew J., Brown, Anthony, Rowland-Jones, Sarah L., Mongkolsapaya, Juthathip, Payne, Rebecca P., Dold, Christina, Lambe, Teresa, Thaventhiran, James E.D., Screaton, Gavin, Barnes, Eleanor, Hopkins, Susan, Hall, Victoria, Duncan, Christopher JA., Richter, Alex, Carroll, Miles, de Silva, Thushan I., Klenerman, Paul, Dunachie, Susanna, and Turtle, Lance
- Abstract
Both infection and vaccination, alone or in combination, generate antibody and T cell responses against SARS-CoV-2. However, the maintenance of such responses – and hence protection from disease – requires careful characterisation. In a large prospective study of UK healthcare workers (Protective immunity from T cells in Healthcare workers (PITCH), within the larger SARS-CoV-2 immunity & reinfection evaluation (SIREN) study) we previously observed that prior infection impacted strongly on subsequent cellular and humoral immunity induced after long and short dosing intervals of BNT162b2 (Pfizer/BioNTech) vaccination.
- Published
- 2023
- Full Text
- View/download PDF
37. The antibody response to SARS-CoV-2 Beta underscores the antigenic distance to other variants.
- Author
-
Liu, Chang, Zhou, Daming, Nutalai, Rungtiwa, Duyvesteyn, Helen M.E., Tuekprakhon, Aekkachai, Ginn, Helen M., Dejnirattisai, Wanwisa, Supasa, Piyada, Mentzer, Alexander J., Wang, Beibei, Case, James Brett, Zhao, Yuguang, Skelly, Donal T., Chen, Rita E., Johnson, Sile Ann, Ritter, Thomas G., Mason, Chris, Malik, Tariq, Temperton, Nigel, and Paterson, Neil G.
- Abstract
Alpha-B.1.1.7, Beta-B.1.351, Gamma-P.1, and Delta-B.1.617.2 variants of SARS-CoV-2 express multiple mutations in the spike protein (S). These may alter the antigenic structure of S, causing escape from natural or vaccine-induced immunity. Beta is particularly difficult to neutralize using serum induced by early pandemic SARS-CoV-2 strains and is most antigenically separated from Delta. To understand this, we generated 674 mAbs from Beta-infected individuals and performed a detailed structure-function analysis of the 27 most potent mAbs: one binding the spike N-terminal domain (NTD), the rest the receptor-binding domain (RBD). Two of these RBD-binding mAbs recognize a neutralizing epitope conserved between SARS-CoV-1 and -2, while 18 target mutated residues in Beta: K417N, E484K, and N501Y. There is a major response to N501Y, including a public IgVH4-39 sequence, with E484K and K417N also targeted. Recognition of these key residues underscores why serum from Beta cases poorly neutralizes early pandemic and Delta viruses. [Display omitted] • Generated 674 antibodies from patients infected with SARS-CoV-2 Beta variant • 18 of 27 most potent mAbs target the 3 mutations in Beta RBD • A major response to N501Y includes a public IgVH4-39 sequence • Two antibodies recognize a neutralizing epitope conserved between SARS-CoV-1 and -2 Liu et al. generated 674 antibodies from patients infected with the SARS-CoV-2 Beta variant. 18 out of 27 most potent neutralizing antibodies isolated target the 3 mutations present in the receptor-binding domain of this variant. This underscores the poor neutralization by Beta serum of early pandemic and Delta viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
38. A delicate balance between antibody evasion and ACE2 affinity for Omicron BA.2.75
- Author
-
Huo, Jiandong, Dijokaite-Guraliuc, Aiste, Liu, Chang, Das, Raksha, Supasa, Piyada, Selvaraj, Muneeswaran, Nutalai, Rungtiwa, Zhou, Daming, Mentzer, Alexander J., Skelly, Donal, Ritter, Thomas G., Amini, Ali, Bibi, Sagida, Adele, Sandra, Johnson, Sile Ann, Paterson, Neil G., Williams, Mark A., Hall, David R., Plowright, Megan, Newman, Thomas A.H., Hornsby, Hailey, de Silva, Thushan I., Temperton, Nigel, Klenerman, Paul, Barnes, Eleanor, Dunachie, Susanna J., Pollard, Andrew J., Lambe, Teresa, Goulder, Philip, Fry, Elizabeth E., Mongkolsapaya, Juthathip, Ren, Jingshan, Stuart, David I., and Screaton, Gavin R.
- Abstract
Variants of SARS CoV-2 have caused successive global waves of infection. These variants, with multiple mutations in the spike protein are thought to facilitate escape from natural and vaccine-induced immunity and often increase in the affinity for ACE2. The latest variant to cause concern is BA.2.75, identified in India where it is now the dominant strain, with evidence of wider dissemination. BA.2.75 is derived from BA.2 and contains four additional mutations in the receptor binding domain (RBD). Here we perform an antigenic and biophysical characterization of BA.2.75, revealing an interesting balance between humoral evasion and ACE2 receptor affinity. ACE2 affinity for BA.2.75 is increased 9-fold compared to BA.2; there is also evidence of escape of BA.2.75 from immune serum, particularly that induced by Delta infection which may explain the rapid spread in India, where BA.2.75 is now the dominant variant. ACE2 affinity appears to be prioritised over greater escape.
- Published
- 2022
- Full Text
- View/download PDF
39. Neutralization of SARS-CoV-2 by Destruction of the Prefusion Spike.
- Author
-
Huo, Jiandong, Zhao, Yuguang, Ren, Jingshan, Zhou, Daming, Duyvesteyn, Helen M.E., Ginn, Helen M., Carrique, Loic, Malinauskas, Tomas, Ruza, Reinis R., Shah, Pranav N.M., Tan, Tiong Kit, Rijal, Pramila, Coombes, Naomi, Bewley, Kevin R., Tree, Julia A., Radecke, Julika, Paterson, Neil G., Supasa, Piyada, Mongkolsapaya, Juthathip, and Screaton, Gavin R.
- Abstract
There are as yet no licensed therapeutics for the COVID-19 pandemic. The causal coronavirus (SARS-CoV-2) binds host cells via a trimeric spike whose receptor binding domain (RBD) recognizes angiotensin-converting enzyme 2, initiating conformational changes that drive membrane fusion. We find that the monoclonal antibody CR3022 binds the RBD tightly, neutralizing SARS-CoV-2, and report the crystal structure at 2.4 Å of the Fab/RBD complex. Some crystals are suitable for screening for entry-blocking inhibitors. The highly conserved, structure-stabilizing CR3022 epitope is inaccessible in the prefusion spike, suggesting that CR3022 binding facilitates conversion to the fusion-incompetent post-fusion state. Cryogenic electron microscopy (cryo-EM) analysis confirms that incubation of spike with CR3022 Fab leads to destruction of the prefusion trimer. Presentation of this cryptic epitope in an RBD-based vaccine might advantageously focus immune responses. Binders at this epitope could be useful therapeutically, possibly in synergy with an antibody that blocks receptor attachment. • CR3022 binds the RBD of SARS-CoV-2 and shows strong neutralization • Neutralization is by destroying the prefusion spike conformation • CR3022 binds a highly conserved epitope that is inaccessible in prefusion spike protein • CR3022 could have therapeutic potential alone or in synergy with a receptor blocker Huo et al. find that the antibody CR3022 binds tightly to the receptor binding domain of the SARS-CoV-2 spike at a site different to that used by the receptor. CR3022 effectively neutralizes the virus, and cryo-EM reveals that it disrupts the spike. Such antibodies could have potential as COVID-19 therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
40. Native-like SARS-CoV-2 Spike Glycoprotein Expressed by ChAdOx1 nCoV-19/AZD1222 Vaccine
- Author
-
Watanabe, Yasunori, Mendonça, Luiza, Allen, Elizabeth R., Howe, Andrew, Lee, Mercede, Allen, Joel D., Chawla, Himanshi, Pulido, David, Donnellan, Francesca, Davies, Hannah, Ulaszewska, Marta, Belij-Rammerstorfer, Sandra, Morris, Susan, Krebs, Anna-Sophia, Dejnirattisai, Wanwisa, Mongkolsapaya, Juthathip, Supasa, Piyada, Screaton, Gavin R., Green, Catherine M., Lambe, Teresa, Zhang, Peijun, Gilbert, Sarah C., and Crispin, Max
- Abstract
Vaccine development against the SARS-CoV-2 virus focuses on the principal target of the neutralizing immune response, the spike (S) glycoprotein. Adenovirus-vectored vaccines offer an effective platform for the delivery of viral antigen, but it is important for the generation of neutralizing antibodies that they produce appropriately processed and assembled viral antigen that mimics that observed on the SARS-CoV-2 virus. Here, we describe the structure, conformation, and glycosylation of the S protein derived from the adenovirus-vectored ChAdOx1 nCoV-19/AZD1222 vaccine. We demonstrate native-like post-translational processing and assembly, and reveal the expression of S proteins on the surface of cells adopting the trimeric prefusion conformation. The data presented here confirm the use of ChAdOx1 adenovirus vectors as a leading platform technology for SARS-CoV-2 vaccines.
- Published
- 2021
- Full Text
- View/download PDF
41. Addressing pandemic-wide systematic errors in the SARS-CoV-2 phylogeny.
- Author
-
Hunt M, Hinrichs AS, Anderson D, Karim L, Dearlove BL, Knaggs J, Constantinides B, Fowler PW, Rodger G, Street T, Lumley S, Webster H, Sanderson T, Ruis C, Kotzen B, de Maio N, Amenga-Etego LN, Amuzu DSY, Avaro M, Awandare GA, Ayivor-Djanie R, Barkham T, Bashton M, Batty EM, Bediako Y, De Belder D, Benedetti E, Bergthaler A, Boers SA, Campos J, Carr RAA, Chen YYC, Cuba F, Dattero ME, Dejnirattisai W, Dilthey A, Duedu KO, Endler L, Engelmann I, Francisco NM, Fuchs J, Gnimpieba EZ, Groc S, Gyamfi J, Heemskerk D, Houwaart T, Hsiao NY, Huska M, Hölzer M, Iranzadeh A, Jarva H, Jeewandara C, Jolly B, Joseph R, Kant R, Ki KKK, Kurkela S, Lappalainen M, Lataretu M, Lemieux J, Liu C, Malavige GN, Mashe T, Mongkolsapaya J, Montes B, Mora JAM, Morang'a CM, Mvula B, Nagarajan N, Nelson A, Ngoi JM, da Paixão JP, Panning M, Poklepovich T, Quashie PK, Ranasinghe D, Russo M, San JE, Sanderson ND, Scaria V, Screaton G, Sessions OM, Sironen T, Sisay A, Smith D, Smura T, Supasa P, Suphavilai C, Swann J, Tegally H, Tegomoh B, Vapalahti O, Walker A, Wilkinson RJ, Williamson C, Zair X, de Oliveira T, Peto TE, Crook D, Corbett-Detig R, and Iqbal Z
- Abstract
The SARS-CoV-2 genome occupies a unique place in infection biology - it is the most highly sequenced genome on earth (making up over 20% of public sequencing datasets) with fine scale information on sampling date and geography, and has been subject to unprecedented intense analysis. As a result, these phylogenetic data are an incredibly valuable resource for science and public health. However, the vast majority of the data was sequenced by tiling amplicons across the full genome, with amplicon schemes that changed over the pandemic as mutations in the viral genome interacted with primer binding sites. In combination with the disparate set of genome assembly workflows and lack of consistent quality control (QC) processes, the current genomes have many systematic errors that have evolved with the virus and amplicon schemes. These errors have significant impacts on the phylogeny, and therefore over the last few years, many thousands of hours of researchers time has been spent in "eyeballing" trees, looking for artefacts, and then patching the tree. Given the huge value of this dataset, we therefore set out to reprocess the complete set of public raw sequence data in a rigorous amplicon-aware manner, and build a cleaner phylogeny. Here we provide a global tree of 4,471,579 samples, built from a consistently assembled set of high quality consensus sequences from all available public data as of June 2024, viewable at https://viridian.taxonium.org. Each genome was constructed using a novel assembly tool called Viridian (https://github.com/iqbal-lab-org/viridian), developed specifically to process amplicon sequence data, eliminating artefactual errors and mask the genome at low quality positions. We provide simulation and empirical validation of the methodology, and quantify the improvement in the phylogeny. We hope the tree, consensus sequences and Viridian will be a valuable resource for researchers., Competing Interests: Conflict of Interest Gavin Screaton sits on the GSK Vaccines Scientific Advisory Board, consults for AstraZeneca, and is a founding member of RQ Biotechnology.
- Published
- 2024
- Full Text
- View/download PDF
42. Concerted deletions eliminate a neutralizing supersite in SARS-CoV-2 BA.2.87.1 spike.
- Author
-
Duyvesteyn HME, Dijokaite-Guraliuc A, Liu C, Supasa P, Kronsteiner B, Jeffery K, Stafford L, Klenerman P, Dunachie SJ, Mongkolsapaya J, Fry EE, Ren J, Stuart DI, and Screaton GR
- Subjects
- Humans, Models, Molecular, Epitopes chemistry, Epitopes immunology, Sequence Deletion, Antibodies, Viral immunology, Binding Sites, Protein Domains, Protein Binding, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus immunology, Spike Glycoprotein, Coronavirus metabolism, SARS-CoV-2 immunology, SARS-CoV-2 genetics, SARS-CoV-2 chemistry, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 virology
- Abstract
BA.2.87.1 represents a major shift in the BA.2 lineage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is unusual in having two lengthy deletions of polypeptide in the spike (S) protein, one of which removes a beta-strand. Here we investigate its neutralization by a variety of sera from infected and vaccinated individuals and determine its spike (S) ectodomain structure. The BA.2.87.1 receptor binding domain (RBD) is structurally conserved and the RBDs are tightly packed in an "all-down" conformation with a small rotation relative to the trimer axis as compared to the closest previously observed conformation. The N-terminal domain (NTD) maintains a remarkably similar structure overall; however, the rearrangements resulting from the deletions essentially destroy the so-called supersite epitope and eliminate one glycan site, while a mutation creates an additional glycan site, effectively shielding another NTD epitope. BA.2.87.1 is relatively easily neutralized but acquisition of additional mutations in the RBD could increase antibody escape allowing it to become a dominant sub-lineage., Competing Interests: Declaration of interests G.R.S. sits on the GSK Vaccines Scientific Advisory Board and is a founder member of RQ Biotechnology. D.I.S. consults for AstraZeneca. Oxford University holds intellectual property related to SARS-CoV-2 mAbs discovered in G.R.S.’s laboratory. S.J.D. is a Scientific Advisor to the Scottish Parliament on COVID-19., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
43. Oligomerization-driven avidity correlates with SARS-CoV-2 cellular binding and inhibition.
- Author
-
Asor R, Olerinyova A, Burnap SA, Kushwah MS, Soltermann F, Rudden LSP, Hensen M, Vasiljevic S, Brun J, Hill M, Chang L, Dejnirattisai W, Supasa P, Mongkolsapaya J, Zhou D, Stuart DI, Screaton GR, Degiacomi MT, Zitzmann N, Benesch JLP, Struwe WB, and Kukura P
- Subjects
- Humans, Virus Internalization drug effects, Antibodies, Viral immunology, Antibodies, Viral metabolism, Thermodynamics, SARS-CoV-2 metabolism, Angiotensin-Converting Enzyme 2 metabolism, Angiotensin-Converting Enzyme 2 chemistry, Spike Glycoprotein, Coronavirus metabolism, Spike Glycoprotein, Coronavirus chemistry, Protein Binding, COVID-19 virology, COVID-19 metabolism, COVID-19 immunology, Protein Multimerization
- Abstract
Cellular processes are controlled by the thermodynamics of the underlying biomolecular interactions. Frequently, structural investigations use one monomeric binding partner, while ensemble measurements of binding affinities generally yield one affinity representative of a 1:1 interaction, despite the majority of the proteome consisting of oligomeric proteins. For example, viral entry and inhibition in SARS-CoV-2 involve a trimeric spike surface protein, a dimeric angiotensin-converting enzyme 2 (ACE2) cell-surface receptor and dimeric antibodies. Here, we reveal that cooperativity correlates with infectivity and inhibition as opposed to 1:1 binding strength. We show that ACE2 oligomerizes spike more strongly for more infectious variants, while exhibiting weaker 1:1 affinity. Furthermore, we find that antibodies use induced oligomerization both as a primary inhibition mechanism and to enhance the effects of receptor-site blocking. Our results suggest that naive affinity measurements are poor predictors of potency, and introduce an antibody-based inhibition mechanism for oligomeric targets. More generally, they point toward a much broader role of induced oligomerization in controlling biomolecular interactions., Competing Interests: Competing interests statement:P.K. is a nonexecutive director, shareholder of and consultant to Refeyn Ltd., J.L.P.B. and W.B.S. are shareholders of and consultants to Refeyn Ltd. W.B.S. and P.K. received the University of Oxford’s COVID-19 Research Response Fund. P.K. has filed a patent for the contrast enhancement methodology and its application to mass measurement of single biomolecules. G.R.S. is on the GSK Vaccines Scientific Advisory Board, a founder shareholder of RQ biotechnology and Jenner investigator. Oxford University holds intellectual property related to the Oxford-AstraZeneca vaccine.
- Published
- 2024
- Full Text
- View/download PDF
44. The alteration of NK cells phenotypes related to the functions and dengue disease outcomes.
- Author
-
Taechasan N, Scherwitzl I, Supasa P, Dejnirattisai W, Sriruksa K, Limpitikul W, Malasit P, Screaton GR, Mongkolsapaya J, and Duangchinda T
- Subjects
- Humans, Child, Male, Female, Interleukin-15 immunology, Lymphocyte Activation, Interleukin-18 immunology, Natural Cytotoxicity Triggering Receptor 3 immunology, Child, Preschool, Dengue immunology, Dengue virology, Severe Dengue immunology, Severe Dengue virology, Adolescent, CD56 Antigen immunology, Interferon Type I immunology, Killer Cells, Natural immunology, Interleukin-12 immunology, Phenotype, Dendritic Cells immunology, Dengue Virus immunology, Interferon-gamma immunology
- Abstract
Natural killer cells (NK cells) are the front line of immune cells to combat pathogens and able to influence the subsequent adaptive immune responses. One of the factors contributing to pathogenesis in dengue hemorrhagic fever (DHF) disease is aberrant immune activation during early phase of infection. This study explored the profile of NK cells in dengue infected pediatric patients with different degrees of disease severity. DHF patients contained higher frequency of activated NK cells but lower ratio of CD56
dim :CD56bright NK subsets. Activated NK cells exhibited alterations in several NK receptors. Interestingly, the frequencies of NKp30 expressing activated NK cells were more pronounced in dengue fever (DF) than in DHF pediatric patients. In vitro functional analysis indicated that degranulation of NK cells in responding to dengue infected dendritic cells (DCs) required cell-cell contact and type I IFNs. Meanwhile, Interferon gamma (IFN-γ) production initially required cell-cell contact and type I IFNs followed by Interleukin-12 (IL-12), Interleukin-15 (IL-15) and Interleukin-18 (IL-18) resulting in the amplification of IFN-γ producing NK cells over time. This study highlighted the complexity and the factors influencing NK cells responses to dengue virus. Degree of activation, phenotypes of activated cells and the crosstalk between NK cells and other immune cells, could modulate the outcome of NK cells function in the dengue disease., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
45. Comparative Analysis of SARS-CoV-2 Antigenicity across Assays and in Human and Animal Model Sera.
- Author
-
Mühlemann B, Wilks SH, Baracco L, Bekliz M, Carreño JM, Corman VM, Davis-Gardner ME, Dejnirattisai W, Diamond MS, Douek DC, Drosten C, Eckerle I, Edara VV, Ellis M, Fouchier RAM, Frieman M, Godbole S, Haagmans B, Halfmann PJ, Henry AR, Jones TC, Katzelnick LC, Kawaoka Y, Kimpel J, Krammer F, Lai L, Liu C, Lusvarghi S, Meyer B, Mongkolsapaya J, Montefiori DC, Mykytyn A, Netzl A, Pollett S, Rössler A, Screaton GR, Shen X, Sigal A, Simon V, Subramanian R, Supasa P, Suthar M, Türeli S, Wang W, Weiss CD, and Smith DJ
- Abstract
The antigenic evolution of SARS-CoV-2 requires ongoing monitoring to judge the immune escape of newly arising variants. A surveillance system necessitates an understanding of differences in neutralization titers measured in different assays and using human and animal sera. We compared 18 datasets generated using human, hamster, and mouse sera, and six different neutralization assays. Titer magnitude was lowest in human, intermediate in hamster, and highest in mouse sera. Fold change, immunodominance patterns and antigenic maps were similar among sera. Most assays yielded similar results, except for differences in fold change in cytopathic effect assays. Not enough data was available for conclusively judging mouse sera, but hamster sera were a consistent surrogate for human first-infection sera., Competing Interests: VMC: Named on patents regarding SARS-CoV-2 serological testing and monoclonal antibodies. MSD: Consultant for Inbios, Vir Biotechnology, Ocugen, Topspin Therapeutics, Moderna, and Immunome. The Diamond laboratory has received unrelated funding support in sponsored research agreements from Moderna, Vir Biotechnology, Generate Biomedicines, and Emergent BioSolutions. YK: Received unrelated funding support from Daiichi Sankyo Pharmaceutical, Toyama Chemical, Tauns Laboratories, Inc., Shionogi & Co. LTD, Otsuka Pharmaceutical, KM Biologics, Kyoritsu Seiyaku, Shinya Corporation, and Fuji Rebio. IE: Research grant and speakers fees from Moderna. BMe: Research grant from Moderna. GRS: Is on the GSK Vaccines Scientific Advisory Board. Oxford University holds intellectual property related to the Oxford-AstraZeneca vaccine. MS: Serves in an advisory role for Ocugen, Inc. SP: Reports that the Uniformed Services University (USU) Infectious Diseases Clinical Research Program (IDCRP), a US Department of Defense institution, and the Henry M. Jackson Foundation (HJF) were funded under a Cooperative Research and Development Agreement to conduct an unrelated phase III COVID-19 monoclonal antibody immunoprophylaxis trial sponsored by AstraZeneca. The HJF, in support of the USU IDCRP, was funded by the Department of Defense Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense to augment the conduct of an unrelated phase III vaccine trial sponsored by AstraZeneca. Both trials were part of the U.S. Government COVID-19 response. Neither is related to the work presented here.
- Published
- 2023
- Full Text
- View/download PDF
46. Generation of SARS-CoV-2 escape mutations by monoclonal antibody therapy.
- Author
-
Ragonnet-Cronin M, Nutalai R, Huo J, Dijokaite-Guraliuc A, Das R, Tuekprakhon A, Supasa P, Liu C, Selvaraj M, Groves N, Hartman H, Ellaby N, Mark Sutton J, Bahar MW, Zhou D, Fry E, Ren J, Brown C, Klenerman P, Dunachie SJ, Mongkolsapaya J, Hopkins S, Chand M, Stuart DI, Screaton GR, and Rokadiya S
- Subjects
- Humans, Antibodies, Monoclonal therapeutic use, Immunotherapy, Mutation, Antibodies, Neutralizing, Antibodies, Viral, SARS-CoV-2 genetics, COVID-19
- Abstract
COVID-19 patients at risk of severe disease may be treated with neutralising monoclonal antibodies (mAbs). To minimise virus escape from neutralisation these are administered as combinations e.g. casirivimab+imdevimab or, for antibodies targeting relatively conserved regions, individually e.g. sotrovimab. Unprecedented genomic surveillance of SARS-CoV-2 in the UK has enabled a genome-first approach to detect emerging drug resistance in Delta and Omicron cases treated with casirivimab+imdevimab and sotrovimab respectively. Mutations occur within the antibody epitopes and for casirivimab+imdevimab multiple mutations are present on contiguous raw reads, simultaneously affecting both components. Using surface plasmon resonance and pseudoviral neutralisation assays we demonstrate these mutations reduce or completely abrogate antibody affinity and neutralising activity, suggesting they are driven by immune evasion. In addition, we show that some mutations also reduce the neutralising activity of vaccine-induced serum., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
47. Corrigendum to "Persistence of immunogenicity after seven COVID-19 vaccines given as third dose boosters following two doses of ChAdOx1 nCov-19 or BNT162b2 in the UK: Three month analyses of the COV-BOOST trial" [J Infect 84(6) (2022) 795-813, 5511].
- Author
-
Liu X, Munro AP, Feng S, Janani L, Aley PK, Babbage G, Baxter D, Bula M, Cathie K, Chatterjee K, Dejnirattisai W, Dodd K, Enever Y, Qureshi E, Goodman AL, Green CA, Harndahl L, Haughney J, Hicks A, van der Klaauw AA, Kwok J, Libri V, Llewelyn MJ, McGregor AC, Minassian AM, Moore P, Mughal M, Mujadidi YF, Holliday K, Osanlou O, Osanlou R, Owens DR, Pacurar M, Palfreeman A, Pan D, Rampling T, Regan K, Saich S, Serafimova T, Saralaya D, Screaton GR, Sharma S, Sheridan R, Sturdy A, Supasa P, Thomson EC, Todd S, Twelves C, Read RC, Charlton S, Hallis B, Ramsay M, Andrews N, Lambe T, Nguyen-Van-Tam JS, Cornelius V, Snape MD, and Faust SN
- Published
- 2023
- Full Text
- View/download PDF
48. Evolution of long-term vaccine-induced and hybrid immunity in healthcare workers after different COVID-19 vaccine regimens.
- Author
-
Moore SC, Kronsteiner B, Longet S, Adele S, Deeks AS, Liu C, Dejnirattisai W, Reyes LS, Meardon N, Faustini S, Al-Taei S, Tipton T, Hering LM, Angyal A, Brown R, Nicols AR, Dobson SL, Supasa P, Tuekprakhon A, Cross A, Tyerman JK, Hornsby H, Grouneva I, Plowright M, Zhang P, Newman TAH, Nell JM, Abraham P, Ali M, Malone T, Neale I, Phillips E, Wilson JD, Murray SM, Zewdie M, Shields A, Horner EC, Booth LH, Stafford L, Bibi S, Wootton DG, Mentzer AJ, Conlon CP, Jeffery K, Matthews PC, Pollard AJ, Brown A, Rowland-Jones SL, Mongkolsapaya J, Payne RP, Dold C, Lambe T, Thaventhiran JED, Screaton G, Barnes E, Hopkins S, Hall V, Duncan CJA, Richter A, Carroll M, de Silva TI, Klenerman P, Dunachie S, and Turtle L
- Subjects
- Humans, COVID-19 Vaccines, BNT162 Vaccine, ChAdOx1 nCoV-19, Prospective Studies, SARS-CoV-2, Antibodies, Neutralizing, Health Personnel, Immunity, Humoral, COVID-19, Vaccines
- Abstract
Background: Both infection and vaccination, alone or in combination, generate antibody and T cell responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the maintenance of such responses-and hence protection from disease-requires careful characterization. In a large prospective study of UK healthcare workers (HCWs) (Protective Immunity from T Cells in Healthcare Workers [PITCH], within the larger SARS-CoV-2 Immunity and Reinfection Evaluation [SIREN] study), we previously observed that prior infection strongly affected subsequent cellular and humoral immunity induced after long and short dosing intervals of BNT162b2 (Pfizer/BioNTech) vaccination., Methods: Here, we report longer follow-up of 684 HCWs in this cohort over 6-9 months following two doses of BNT162b2 or AZD1222 (Oxford/AstraZeneca) vaccination and up to 6 months following a subsequent mRNA booster vaccination., Findings: We make three observations: first, the dynamics of humoral and cellular responses differ; binding and neutralizing antibodies declined, whereas T and memory B cell responses were maintained after the second vaccine dose. Second, vaccine boosting restored immunoglobulin (Ig) G levels; broadened neutralizing activity against variants of concern, including Omicron BA.1, BA.2, and BA.5; and boosted T cell responses above the 6-month level after dose 2. Third, prior infection maintained its impact driving larger and broader T cell responses compared with never-infected people, a feature maintained until 6 months after the third dose., Conclusions: Broadly cross-reactive T cell responses are well maintained over time-especially in those with combined vaccine and infection-induced immunity ("hybrid" immunity)-and may contribute to continued protection against severe disease., Funding: Department for Health and Social Care, Medical Research Council., Competing Interests: Declaration of interests S.J.D. is a Scientific Advisor to the Scottish Parliament on COVID-19, for which she receives a fee. A.J.P. is Chair of UK Department of Health and Social Care’s (DHSC) Joint Committee on Vaccination and Immunisation (JCVI) but does not participate in policy decisions on COVID-19 vaccines. He was previously a member of the WHO’s SAGE. The views expressed in this article do not necessarily represent the views of DHSC, JCVI, or WHO. A.J.P. is chief investigator on clinical trials of Oxford University’s COVID-19 vaccine funded by NIHR. Oxford University has entered a joint COVID-19 vaccine development partnership with AstraZeneca. G.S. sits on the GSK Vaccines Scientific Advisory Board and is a founder member of RQ Biotechnology., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
49. A delicate balance between antibody evasion and ACE2 affinity for Omicron BA.2.75.
- Author
-
Huo J, Dijokaite-Guraliuc A, Liu C, Zhou D, Ginn HM, Das R, Supasa P, Selvaraj M, Nutalai R, Tuekprakhon A, Duyvesteyn HME, Mentzer AJ, Skelly D, Ritter TG, Amini A, Bibi S, Adele S, Johnson SA, Paterson NG, Williams MA, Hall DR, Plowright M, Newman TAH, Hornsby H, de Silva TI, Temperton N, Klenerman P, Barnes E, Dunachie SJ, Pollard AJ, Lambe T, Goulder P, Fry EE, Mongkolsapaya J, Ren J, Stuart DI, and Screaton GR
- Subjects
- Humans, Angiotensin-Converting Enzyme 2, SARS-CoV-2, Antibodies, COVID-19, Hepatitis D
- Abstract
Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have caused successive global waves of infection. These variants, with multiple mutations in the spike protein, are thought to facilitate escape from natural and vaccine-induced immunity and often increase in affinity for ACE2. The latest variant to cause concern is BA.2.75, identified in India where it is now the dominant strain, with evidence of wider dissemination. BA.2.75 is derived from BA.2 and contains four additional mutations in the receptor-binding domain (RBD). Here, we perform an antigenic and biophysical characterization of BA.2.75, revealing an interesting balance between humoral evasion and ACE2 receptor affinity. ACE2 affinity for BA.2.75 is increased 9-fold compared with BA.2; there is also evidence of escape of BA.2.75 from immune serum, particularly that induced by Delta infection, which may explain the rapid spread in India, where where there is a high background of Delta infection. ACE2 affinity appears to be prioritized over greater escape., Competing Interests: Declaration of interests G.R.S. sits on the GSK Vaccines Scientific Advisory Board, consults for Astra Zeneca, and is a founding member of RQ Biotechnology. Oxford University holds intellectual property related to the Oxford-Astra Zeneca vaccine and SARS-CoV-2 mAbs discovered in G.R.S.’s laboratory. A.J.P. is Chair of UK Dept. Health and Social Care’s (DHSC) Joint Committee on Vaccination & Immunisation (JCVI) but does not participate in the JCVI COVID-19 committee and is a member of the WHO’s SAGE. The views expressed in this article do not necessarily represent the views of DHSC, JCVI, or WHO. The University of Oxford has entered into a partnership with AstraZeneca on coronavirus vaccine development. T.L. is named as an inventor on a patent application covering this SARS-CoV-2 vaccine and was a consultant to Vaccitech for an unrelated project whilst the study was conducted. S.J.D. is a scientific advisor to the Scottish Parliament on COVID-19., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
50. A group of infection-enhancing and focus size-reducing monoclonal antibodies recognized an 'a and c' strands epitope in the pr domain of Dengue Virus prM.
- Author
-
Keelapang P, Supasa P, Sriburi R, Puttikhunt C, Cardosa J, Kasinrerk W, Malasit P, and Sittisombut N
- Abstract
Partial cleavage of a dengue virus envelope protein, prM, by furin results in a mixture of extracellular particles with variable levels of maturation and infectivity. Partially mature particles can infect leukocytes via interaction between the prM-anti-prM antibody complex with Fcγ receptors. Known prM epitopes involved in antibody-mediated infection are localized to the pr domain. In this study, a group of murine anti-prM monoclonal antibodies with strong infection-enhancing activity was found to reduce the focus size of subsets of multiple dengue serotypes that they could enhance. By employing sets of overlapping peptides, four antibodies recognizing 2-mercaptoethanol-insensitive epitopes were mapped to a common tetrapeptide located distantly in the b-c loop and furin binding site. Substitution mutations of each, or both, of the tetrapeptides in virus-like particles, however, failed to reduce binding. Further mapping experiments were performed using immature virus-like particles with abolished furin binding site to minimize the differential influence of various pr substitutions on pr-M cleavage. Reduction of antibody binding was detected when single alanine substitutions were introduced into the 'a' strand and 'c' strand of pr domain. These findings suggest that the pr 'a and c' strands region is the major binding site of these unusual focus size-reducing anti-prM antibodies., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.