1
|
Quirk GE, Schoenle MV, Peyton KL, Uhrlaub JL, Lau B, Liang CY, Burgess JL, Ellingson K, Beitel S, Romine J, Lutrick K, Fowlkes A, Britton A, Tyner HL, Caban-Martinez AJ, Naleway A, Gaglani M, Yoon S, Edwards LJ, Olsho L, Dake M, Valdez R, Gordon A, Diamond MS, LaFleur BJ, Nikolich JŽ, Sprissler R, Worobey M, Bhattacharya D. Intrinsic immunogenicity is a major determinant of type-specific responses in SARS-CoV-2 infections. Nat Immunol 2025:10.1038/s41590-025-02162-2. [PMID: 40425779 DOI: 10.1038/s41590-025-02162-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/11/2025] [Indexed: 05/29/2025]
Abstract
Few type-specific antibodies that recognize drifted epitopes are made during post-vaccination exposures to SARS-CoV-2 variants1-12, perhaps due to suppression by previous immunity. We compared type-specific B cell responses in unvaccinated and vaccinated individuals with Delta and Omicron BA.1 SARS-CoV-2 variant infections. For both Delta, which is antigenically similar to the vaccine strain, and the more distant BA.1 variant, neutralizing antibodies were greater in post-vaccination variant infections than in primary variant infections. Delta type-specific memory B cells were reduced in post-vaccination Delta infections relative to primary variant infections. Yet some drifted epitopes in the Delta variant elicited minimal responses even in primary infections. For BA.1 infections, type-specific antibodies and memory B cells were mostly undetectable, irrespective of previous immunity. Thus, poor intrinsic antigenicity of drifted epitopes in Delta and BA.1 infections superseded the impact of previous immunity. Enhancing the immunogenicity of vaccine antigens may promote type-specific responses.
Collapse
Affiliation(s)
- Grace E Quirk
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
| | - Marta V Schoenle
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Kameron L Peyton
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Branden Lau
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Chieh-Yu Liang
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Jefferey L Burgess
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Katherine Ellingson
- Department of Epidemiology and Biostatistics, Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Shawn Beitel
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - James Romine
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Karen Lutrick
- College of Medicine-Tucson, University of Arizona, Tucson, AZ, USA
| | - Ashley Fowlkes
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Amadea Britton
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Alberto J Caban-Martinez
- Department of Public Health Services, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allison Naleway
- Kaiser Permanente Northwest Center for Health Research, Portland, OR, USA
| | | | - Sarang Yoon
- Rocky Mountain Center for Occupational and Environmental Health, Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, UT, USA
| | | | | | - Michael Dake
- Office of the Senior Vice-President for Health Sciences, University of Arizona, Tucson, AZ, USA
| | - Riccardo Valdez
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Bonnie J LaFleur
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Ryan Sprissler
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Michael Worobey
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA.
- BIO5 Institute, University of Arizona, Tucson, AZ, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
2
|
You L, Zhang L, Ouyang S, Gao B, Li Y, Li J, Wu N, Wang H, Sun S, Li J, Yin Z, Xu Z, Chen Y, Zhu Y, Zhang S, Xu Z, Zhang T, Liu Z, Huang C, Li B, Qu J, Su B, Yeap LS. Multiple infections with Omicron variants increase breadth and potency of Omicron-specific neutralizing antibodies. Cell Discov 2025; 11:49. [PMID: 40389403 PMCID: PMC12089387 DOI: 10.1038/s41421-025-00800-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/07/2025] [Indexed: 05/21/2025] Open
Abstract
Despite high vaccination rates, highly evolved Omicron variants have caused widespread infections and, in some cases, recurrent infections in the human population. As the population continues to be threatened by new variants, it is critical to understand how the dynamic cross-reactive antibody response evolves and affects protection. Here, we longitudinally profiled neutralizing antibodies in individuals who experienced three Omicron waves in China over an 18-month period following the lifting of the COVID restriction. We found that individuals with BA.5/BF.7 and XBB dual infections had increased breadth and neutralizing potency of Omicron-specific antibodies compared to those with a BA.5/BF.7 single infection, and were thus more resistant to JN.1/XDV.1 infection in the third wave. During the second infection, a new imprint based on the previously infected variant was established, and the antibodies developed high cross-reactivity against the Omicron variants and less against vaccine-derived WT SARS-CoV-2. Our results suggest that the high titer and breadth of cross-reactive antibodies from multiple infections may be protective against future infection with Omicron variants such as JN.1, but may still be vulnerable to antigenically advanced subvariants such as KP.3.1.1 and XEC.
Collapse
Affiliation(s)
- Lei You
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luning Zhang
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengqun Ouyang
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Gao
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanan Li
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Jialu Li
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningbo Wu
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Wang
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiqi Sun
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinfeng Li
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi Yin
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyang Xu
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Chen
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Zhu
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyan Zhang
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhan Xu
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyu Zhang
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanxin Huang
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieming Qu
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, China.
- National Research Center for Translational Medicine at Shanghai, Shanghai, China.
| | - Bing Su
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Leng-Siew Yeap
- Shanghai Institute of Immunology and Department of Pulmonary and Critical Care Medicine, Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Wrynla XH, Bates TA, Trank-Greene M, Wahedi M, Hinchliff A, Curlin ME, Tafesse FG. Immune imprinting and vaccine interval determine antibody responses to monovalent XBB.1.5 COVID-19 vaccination. COMMUNICATIONS MEDICINE 2025; 5:182. [PMID: 40382525 PMCID: PMC12085693 DOI: 10.1038/s43856-025-00898-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND As COVID-19 becomes endemic and vaccines are annually adapted, exposure intervals and immune imprinting become critical considerations for vaccination strategy. Imprinting by the ancestral spike protein affected bivalent Wuhan-Hu-1/BA.4-5 vaccine responses. We assess the persistence of imprinting in antibody responses to the more recent XBB.1.5 monovalent formulation. METHODS We quantified live virus-neutralizing antibodies by focus reduction neutralization test and ancestral spike receptor-binding isotype titers by immunosorbent assay in individuals before and after XBB.1.5 vaccination. We compared responses between those who previously received three to four doses of Wuhan-Hu-1 vaccine and one dose of bivalent Wuhan-Hu-1/BA.4-5 (bivalent recipients) and those who received three to four doses of Wuhan-Hu-1 (bivalent non-recipients). RESULTS We report that before XBB.1.5 vaccination, bivalent non-recipients have decreased breadth and potency of neutralization. At post-vaccination, non-recipients exhibit greater boosting of neutralizing antibodies against XBB.1.5 (18.4X versus 6.2X), EG.5.1 (30.9X versus 7.0X), and JN.1 (9.2X versus 3.7X) variants with comparable breadth and trends toward greater potency. Greater boosting in non-recipients is similarly observed for spike-binding IgA and total IgG/A/M but not IgG nor IgM. Bivalent non-recipients had longer intervals between vaccination, which may enhance antibody responses; however, bivalent receipt and interval are tightly linked, preventing isolation of individual contributions to boosting. Nonetheless, back-boosting of ancestral SARS-CoV-2 titers in both participant groups provides interval-independent evidence that imprinting persists. CONCLUSIONS Our findings indicate that immune imprinting continues to affect humoral immunity elicited by the XBB.1.5 vaccine. Both imprinting and exposure intervals are important phenomena underlying immunogenicity of future variant-adapted COVID-19 vaccines.
Collapse
Affiliation(s)
- Xammy Huu Wrynla
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Timothy A Bates
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Mila Trank-Greene
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Mastura Wahedi
- Department of Occupational Health, Oregon Health and Science University, Portland, OR, USA
| | - Audrey Hinchliff
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Marcel E Curlin
- Department of Occupational Health, Oregon Health and Science University, Portland, OR, USA.
- Division of Infectious Diseases, Oregon Health and Science University, Portland, OR, USA.
| | - Fikadu G Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
4
|
Kato H, Sano K, Miyakawa K, Kurosawa T, Horikawa K, Kimura Y, Goto A, Ryo A. Humoral and Cell-Mediated Immunity Against SARS-CoV-2 in Healthcare Personnel Who Received Multiple mRNA Vaccines: A 4-Year Observational Study. Infect Dis Rep 2025; 17:42. [PMID: 40407644 PMCID: PMC12101388 DOI: 10.3390/idr17030042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/26/2025] Open
Abstract
Background/Objectives: The long-term effects of multiple updated vaccinations against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) have not been clarified. Humoral or cellular immunity dynamics in healthcare workers for four years were analyzed. Methods: Blood samples were collected at five time points from April 2021 to January 2024. Humoral immunity was analyzed using the 50% neutralizing titer (NT50) against the original Omicron XBB and Omicron BA.2.86 strains and cellular immunity were analyzed using the ELISpot interferon-gamma releasing assay. NT50s and the spot-forming count (SFC) of the ELISpot assay were compared in the SARS-CoV-2 Omicron XBB-, Omicron-infected, and uninfected subjects. Results: 32 healthcare workers (median age, 47 years) who received 3-7 vaccine doses were enrolled. The NT50s against the original strain decreased after the second vaccination but were maintained after the third vaccine dose. NT50s against the Omicron XBB and BA.2.86 strains were detected before the Omicron vaccine was introduced and increased following the updated vaccination. The NT50s against the Omicron XBB and BA.2.86 strains were elevated after natural infection by the Omicron strain, albeit without differences compared with the findings in uninfected subjects. Multivariate regression analysis revealed no confounder that affected the antibody titer against the BA.2.86 strain at the fifth blood sampling. The median number of SFCs ranged from 78 to 208 after the first two doses. Conclusions: Multiple vaccinations induced the production of antibodies with divergent activity against emerging mutant strains and enhanced protective effects against the original strain. This finding supported the importance of updated vaccination.
Collapse
Affiliation(s)
- Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Yokohama 236-0004, Japan
- Emerging Infectious Diseases Research Center, Yokohama City University Hospital, Yokohama 236-0004, Japan
| | - Kaori Sano
- Influenza Research Center, National Institute of Infectious Diseases, Japan Institute for Health Security, Musashimurayama 208-0011, Japan
| | - Kei Miyakawa
- Influenza Research Center, National Institute of Infectious Diseases, Japan Institute for Health Security, Musashimurayama 208-0011, Japan
| | - Takayuki Kurosawa
- Clinical Laboratory Department, Yokohama City University Hospital, Yokohama 236-0004, Japan
| | - Kazuo Horikawa
- Emerging Infectious Diseases Research Center, Yokohama City University Hospital, Yokohama 236-0004, Japan
- Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan
| | - Yayoi Kimura
- Emerging Infectious Diseases Research Center, Yokohama City University Hospital, Yokohama 236-0004, Japan
- Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan
| | - Atsushi Goto
- Emerging Infectious Diseases Research Center, Yokohama City University Hospital, Yokohama 236-0004, Japan
- Department of Public Health, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Akihide Ryo
- Department of Bioinformatics and Integrative Omics, National Institute of Infectious Diseases, Japan Institute for Health Security, Musashimurayama 208-0011, Japan
| |
Collapse
|
5
|
Dadonaite B, Burrell AR, Logue J, Chu HY, Payne DC, Haslam DB, Staat MA, Bloom JD. SARS-CoV-2 neutralizing antibody specificities differ dramatically between recently infected infants and immune-imprinted individuals. J Virol 2025; 99:e0010925. [PMID: 40130874 PMCID: PMC11998527 DOI: 10.1128/jvi.00109-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
The immune response to viral infection is shaped by past exposures to related virus strains, a phenomenon known as imprinting. For severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), much of the population has been imprinted by a viral spike from an early strain, either through vaccination or infection during the early stages of the COVID-19 pandemic. As a consequence of this imprinting, infection with more recent SARS-CoV-2 strains primarily boosts cross-reactive antibodies elicited by the imprinting strain. Here we compare the neutralizing antibody specificities of imprinted individuals versus infants infected with a recent strain. Specifically, we use pseudovirus-based deep mutational scanning to measure how spike mutations affect neutralization by the serum antibodies of adults and children imprinted by the original vaccine versus infants with a primary infection by an XBB* variant. While the serum neutralizing activity of the imprinted individuals primarily targets the spike receptor-binding domain (RBD), the serum neutralizing activity of infants infected with only XBB* mostly targets the spike N-terminal domain. In these infants, secondary exposure to the XBB* spike via vaccination shifts more of the neutralizing activity toward the RBD, although the specific RBD sites targeted are different from imprinted adults. The dramatic differences in neutralization specificities among individuals with different exposure histories likely impact SARS-CoV-2 evolution.IMPORTANCEWe show that a person's exposure history to different SARS-CoV-2 strains strongly affects which regions on the viral spike that their neutralizing antibodies target. In particular, infants who have just been infected once with a recent viral strain make neutralizing antibodies that target different regions of the viral spike than adults or children who have been exposed to both older and more recent strains. This person-to-person heterogeneity means that the same viral mutation can have different impacts on the antibody immunity of different people.
Collapse
Affiliation(s)
- Bernadeta Dadonaite
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Allison R. Burrell
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jenni Logue
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Helen Y. Chu
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Daniel C. Payne
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - David B. Haslam
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mary A. Staat
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Howard Hughes Medical Institute, Seattle, Washington, USA
| |
Collapse
|
6
|
Carreño JM, Lerman B, Singh G, Abbad A, Yellin T, Ehrenhaus J, Fried M, Nardulli JR, Kang HM, Mulder LCF, Gleason C, Srivastava K, PVI study group, Simon V, Krammer F. XBB.1.5 monovalent vaccine induces lasting cross-reactive responses to SARS-CoV-2 variants such as HV.1 and JN.1, as well as SARS-CoV-1, but elicits limited XBB.1.5 specific antibodies. mBio 2025; 16:e0360724. [PMID: 40042313 PMCID: PMC11980561 DOI: 10.1128/mbio.03607-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/13/2025] [Indexed: 04/10/2025] Open
Abstract
The evolution of the antibody response to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is impacted by the nature and number of antigenic exposures. First-generation coronavirus disease 2019 (COVID-19) vaccines encoded an ancestral spike protein. Updated bivalent vaccines and breakthrough infections have shaped the intricate diversity of the polyclonal antibody response and specificity of individual antibody clones. We and others previously showed that bivalent vaccines containing the ancestral and Omicron (BA.5) spikes induce high levels of cross-reactive antibodies but undetectable BA.5-specific antibodies in serum. Here, we assessed sera collected before as well as 1 and 3 months following administration of an updated XBB.1.5 monovalent vaccine to individuals with diverse infection and vaccination histories. Vaccination increased neutralization against recent variants of concern, including HV.1, JN.1, and the vaccine-homologous XBB.1.5. Antibody binding and avidity against ancestral and XBB.1.5 antigens significantly increased after vaccination. However, antibody depletion experiments showed that most of the response was cross-reactive to the ancestral spike, and only low levels of XBB.1.5-specific antibodies to the spike or the receptor-binding domain were detected. Importantly, increased antibody levels were still detectable in circulation 3 months post-vaccination and cross-reacted with severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) as measured by pseudovirus neutralization and binding assays. Overall, our data suggest that the XBB.1.5 monovalent vaccine predominantly elicits a cross-reactive response imprinted by viral spike antigens encountered early during the pandemic.IMPORTANCEUpdated COVID-19 vaccine formulations and SARS-CoV-2 exposure history affect the antibody response to SARS-CoV-2. High titers of antibodies are induced in serum by XBB.1.5 monovalent vaccination. Antibody depletion experiments reveal that the majority of the antibody response is cross-reactive to the ancestral spike, despite vaccination increasing neutralization against recently circulating Omicron variants. Vaccine-induced SARS-CoV-2 antibodies cross-react with SARS-CoV-1 and remain in the bloodstream for at least 3 months after immunization.
Collapse
Affiliation(s)
- Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Brian Lerman
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Anass Abbad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Temima Yellin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jordan Ehrenhaus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Miriam Fried
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jessica R. Nardulli
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Hyun Min Kang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Lubbertus C. F. Mulder
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Charles Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
| | - PVI study group
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Purushotham JN, Lutz HL, Parker E, Andersen KG. Immunological drivers of zoonotic virus emergence, evolution, and endemicity. Immunity 2025; 58:784-796. [PMID: 40168990 PMCID: PMC11981831 DOI: 10.1016/j.immuni.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025]
Abstract
The disruption of natural ecosystems caused by climate change and human activity is amplifying the risk of zoonotic spillover, presenting a growing global health threat. In the past two decades, the emergence of multiple zoonotic viruses has exposed critical gaps in our ability to predict epidemic trajectories and implement effective interventions. RNA viruses, in particular, are challenging to control due to their high mutation rates and ability to adapt and evade immune defenses. To better prepare for future outbreaks, it is vital that we deepen our understanding of the factors driving viral emergence, transmission, and persistence in human populations. Specifically, deciphering the interactions between antibody-mediated immunity and viral evolution will be key. In this perspective, we explore these dynamic relationships and highlight research priorities that may guide the development of more effective strategies to mitigate the impact of emerging infectious diseases.
Collapse
Affiliation(s)
- Jyothi N Purushotham
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - Holly L Lutz
- Denver Museum of Nature and Science, Denver, CO, USA
| | - Edyth Parker
- The Institute of Genomics and Global Health (IGH), Redeemer's University, Ede, Osun, Nigeria
| | - Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA.
| |
Collapse
|
8
|
Atti A, England A, Sung J, Foulkes S, Islam J, Otter A, Thomas K, Hallis B, Hopkins S, Charlton S, Hall V. Estimating neutralising antibody responses against emerging SARS-CoV-2 variants utilising convalescent sera before the roll-out of XBB-lineage vaccines. Vaccine 2025; 51:126898. [PMID: 39983539 DOI: 10.1016/j.vaccine.2025.126898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND SARS-CoV-2 immune escape variants can alter existing vaccine effectiveness. In September 2023, we aimed to predict the neutralising response against BA.2.86 offered by XBB-lineage vaccines before vaccine roll-out utilising XBB.1.5 convalescent sera. We then assessed the response to XBB-lineage boosters from different individuals in the same cohort. METHODS A total of 78 sera samples (pre/post-XBB.1.5 infection and pre/post-XBB-lineage vaccination) were tested for live microneutralisation against SARS-CoV-2 Victoria and Omicron subvariants. Geometric means (GM) of neutralising antibody titres (nAbT) pre- and post-infection/vaccination were compared. RESULTS After XBB.1.5 infection, a 4-fold increase in neutralising antibody titres against BA.2.86 was observed compared to pre-infection titres (GM 51 vs 210, p ≤ 0.0001). A similar increase in BA.2.86 nAbT was seen post-XBB-lineage vaccination (GM 144 vs 600, fold change = 4.17, p ≤ 0.0001). CONCLUSION XBB.1.5 infection was a suitable proxy to predict the neutralisation response following XBB-lineage vaccination. Our findings may support future vaccine development and vaccination strategies.
Collapse
Affiliation(s)
- Ana Atti
- United Kingdom Health Security Agency (UKHSA), 10 South Colonnade, Canary Wharf, London E14 4PU, United Kingdom.
| | - Anna England
- Vaccine Development and Evaluation Centre (VDEC), UK Health Security Agency (UKHSA), Manor Farm Road, Porton, Salisbury SP4 0JG, United Kingdom
| | - Julia Sung
- Vaccine Development and Evaluation Centre (VDEC), UK Health Security Agency (UKHSA), Manor Farm Road, Porton, Salisbury SP4 0JG, United Kingdom
| | - Sarah Foulkes
- United Kingdom Health Security Agency (UKHSA), 10 South Colonnade, Canary Wharf, London E14 4PU, United Kingdom
| | - Jasmin Islam
- United Kingdom Health Security Agency (UKHSA), 10 South Colonnade, Canary Wharf, London E14 4PU, United Kingdom
| | - Ashley Otter
- Emerging Pathogen Serology, UK Health Security Agency (UKHSA), Manor Farm Road, Porton, Salisbury SP4 0JG, United Kingdom
| | - Kelly Thomas
- Vaccine Development and Evaluation Centre (VDEC), UK Health Security Agency (UKHSA), Manor Farm Road, Porton, Salisbury SP4 0JG, United Kingdom
| | - Bassam Hallis
- Vaccine Development and Evaluation Centre (VDEC), UK Health Security Agency (UKHSA), Manor Farm Road, Porton, Salisbury SP4 0JG, United Kingdom
| | - Susan Hopkins
- United Kingdom Health Security Agency (UKHSA), 10 South Colonnade, Canary Wharf, London E14 4PU, United Kingdom
| | - Sue Charlton
- Vaccine Development and Evaluation Centre (VDEC), UK Health Security Agency (UKHSA), Manor Farm Road, Porton, Salisbury SP4 0JG, United Kingdom
| | - Victoria Hall
- United Kingdom Health Security Agency (UKHSA), 10 South Colonnade, Canary Wharf, London E14 4PU, United Kingdom
| |
Collapse
|
9
|
Case JB, Jain S, Suthar MS, Diamond MS. SARS-CoV-2: The Interplay Between Evolution and Host Immunity. Annu Rev Immunol 2025; 43:29-55. [PMID: 39705164 DOI: 10.1146/annurev-immunol-083122-043054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
The persistence of SARS-CoV-2 infections at a global level reflects the repeated emergence of variant strains encoding unique constellations of mutations. These variants have been generated principally because of a dynamic host immune landscape, the countermeasures deployed to combat disease, and selection for enhanced infection of the upper airway and respiratory transmission. The resulting viral diversity creates a challenge for vaccination efforts to maintain efficacy, especially regarding humoral aspects of protection. Here, we review our understanding of how SARS-CoV-2 has evolved during the pandemic, the immune mechanisms that confer protection, and the impact viral evolution has had on transmissibility and adaptive immunity elicited by natural infection and/or vaccination. Evidence suggests that SARS-CoV-2 evolution initially selected variants with increased transmissibility but currently is driven by immune escape. The virus likely will continue to drift to maintain fitness until countermeasures capable of disrupting transmission cycles become widely available.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Shilpi Jain
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael S Diamond
- Department of Pathology & Immunology; Department of Molecular Microbiology; and Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
10
|
Maltseva M, Galipeau Y, McCluskie P, Castonguay N, Cooper CL, Langlois MA. Systemic and Mucosal Antibody Responses to SARS-CoV-2 Variant-Specific Prime-and-Boost and Prime-and-Spike Vaccination: A Comparison of Intramuscular and Intranasal Bivalent Vaccine Administration in a Murine Model. Vaccines (Basel) 2025; 13:351. [PMID: 40333249 PMCID: PMC12031244 DOI: 10.3390/vaccines13040351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 05/09/2025] Open
Abstract
Background: The rapid genetic evolution of SARS-CoV-2 has led to the emergence of immune-evading, highly transmissible variants of concern (VOCs). This prompts the need for next-generation vaccines that elicit robust mucosal immunity in the airways to directly curb viral infection. Objective: Here, we investigate the impact of heterologous variant prime-boost regimens on humoral responses, focusing on intramuscular (IM) and intranasal (IN) routes of administration. Using a murine model, we assessed the immunogenicity of unadjuvanted protein boosts with Wu-1, Omicron BA.4/5, or Wu-1 + BA.4/5 spike antigens following monovalent or bivalent IM priming with mRNA-LNP vaccines. Results: IM priming induced strong systemic total and neutralizing antibody responses that were further enhanced by IN boosts with BA.4/5. IN boosting achieved the broadest serum neutralization across all VOCs tested. Notably, bivalent mRNA-LNP IM priming induced robust, cross-variant serum neutralizing antibody production, independent of subsequent IN boost combinations. Conclusions: Our findings highlight the benefit of including distinct antigenic variants in the prime vaccination followed by a variant-tailored IN boost to elicit both systemic and mucosal variant-specific responses that are potentially capable of reducing SARS-CoV-2 transmission.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Pauline McCluskie
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nicolas Castonguay
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Curtis L. Cooper
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
11
|
Sop J, Mercado A, Figueroa A, Beckey TP, Traut CC, Zhang L, Smith KN, Blankson JN. The XBB.1.5 mRNA booster vaccine does not significantly increase the percentage of XBB.1.5 mono-reactive T cells. Front Immunol 2025; 16:1513175. [PMID: 40145092 PMCID: PMC11936820 DOI: 10.3389/fimmu.2025.1513175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Recent efforts in vaccine development have targeted spike proteins from evolving SARS-CoV-2 variants. In this study, we analyzed T cell responses to the XBB.1.5 and BA.2.86 subvariants in individuals who previously received bivalent vaccines containing mRNA for ancestral and BA.5 spike proteins. T cell-mediated cytokine responses to spike proteins from both variants were largely preserved. To determine the mechanism of this preserved recognition, we utilized the functional expansion of specific T cells (FEST) assay to distinguish between the presence of T cells that cross-recognized ancestral and variant epitopes versus distinct populations of T cells that were mono-reactive for ancestral or variant epitopes. We found the majority of spike-specific T cells cross-recognized the ancestral spike and the XBB.1.5 and BA.2.86 subvariants, with less than 10% of T cells being mono-reactive for either variant. Interestingly, immunization with the XBB.1.5 monovalent booster vaccine did not significantly increase the percentage of XBB.1.5 mono-reactive T cells. Our results suggest a potential limitation in the induction of mono-reactive T cell responses by variant-specific booster vaccines.
Collapse
Affiliation(s)
- Joel Sop
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Alicia Mercado
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Alexis Figueroa
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Tyler P. Beckey
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Caroline C. Traut
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Li Zhang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Kellie N. Smith
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Joel N. Blankson
- Department of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Ciabattini A, Pettini E, Fiorino F, Polvere J, Lucchesi S, Coppola C, Costagli S, Pastore G, Sicuranza A, Tozzi M, Lippi A, Panza F, Bocchia M, Bucalossi A, Garosi G, Bennett D, Bernazzali S, Fabbiani M, Montagnani F, Medaglini D. Longitudinal immunogenicity cohort study of SARS-CoV-2 mRNA vaccines across individuals with different immunocompromising conditions: heterogeneity in the immune response and crucial role of Omicron-adapted booster doses. EBioMedicine 2025; 113:105577. [PMID: 39908650 PMCID: PMC11847306 DOI: 10.1016/j.ebiom.2025.105577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Individuals with primary and secondary immunodeficiencies, being more susceptible to infections, are a priority for vaccination. Here, we determined and compared in a longitudinal study the immune response elicited by SARS-CoV-2 vaccination across different groups of individuals who are immunocompromised. METHODS In the PatoVac_COV longitudinal prospective single-centre study, the spike-specific B cell and antibody responses to SARS-CoV-2 mRNA vaccination were compared across 5 different groups of individuals with haematological malignancies, hematopoietic stem cell (HCT) or solid organ transplantation (SOT), undergoing haemodialysis, and people living with HIV (PLWH), for a total of 585 participants. Data from participants who were immunocompromised were compared to a group of 123 participants who were immunocompetent. Blood samples were collected before and after each vaccine administration, up to 2 years. FINDINGS A different immune responsiveness was observed after the first two vaccine doses, with haematological, haemodialysis, and SOT participants showing reduced responsiveness compared to HCT and PLWH, and relative to the comparison group. Spike-specific B cell response was both slower and lower in all groups except in PLWH when compared to participants who were immunocompetent. However, the first booster dose enhanced both the B and the antibody responses in all groups, that persisted up to 2 years after the first vaccine administration. The administration of Omicron-adapted booster vaccines promoted a primary BA.2 RBD-specific B cell response, especially in participants who were immunocompromised. Despite repeated vaccinations, a subset of persistent low-responders, especially among SOT, was identified. INTERPRETATION Our study highlights the heterogeneous immune response across individuals with different pathologies, the pivotal role of the first booster dose, the primary activation of Omicron-specific B cells elicited by updated variant-adapted vaccines and the persistence of low-responders despite multiple vaccine administrations. These aspects have a clinical relevance for planning vaccination schedules tailored for individuals with different immunocompromising conditions. FUNDING This work was supported by funds from the Department of Medical Biotechnologies of the University of Siena, and from EU within the NextGenerationEU-MUR PNRR Tuscany Health Ecosystem (Project no ECS00000017-THE).
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Department of Medicine and Surgery, LUM University "Giuseppe Degennaro", Bari, Italy
| | - Jacopo Polvere
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Chiara Coppola
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Simone Costagli
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Anna Sicuranza
- Haematology Unit, Department of Medical Science, Surgery and Neuroscience, University Hospital of Siena, Siena, Italy
| | - Monica Tozzi
- Cellular Therapy Unit, Department of Innovation, Experimentation, Clinical and Translational Research, University Hospital of Siena, Siena, Italy
| | - Arianna Lippi
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Panza
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Monica Bocchia
- Haematology Unit, Department of Medical Science, Surgery and Neuroscience, University Hospital of Siena, Siena, Italy
| | - Alessandro Bucalossi
- Cellular Therapy Unit, Department of Innovation, Experimentation, Clinical and Translational Research, University Hospital of Siena, Siena, Italy
| | - Guido Garosi
- Department of Medical Science, Nephrology, Dialysis and Transplantation Unit, University Hospital of Siena, Italy
| | - David Bennett
- Respiratory Disease and Lung Transplant Unit, University of Siena, University Hospital of Siena, Italy
| | - Sonia Bernazzali
- Department of Cardiac Surgery, University of Siena, Siena, Italy
| | - Massimiliano Fabbiani
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Montagnani
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
13
|
Ioannou GN, Berry K, Rajeevan N, Li Y, Yan L, Huang Y, Lin HM, Bui D, Hynes DM, Rowneki M, Bohnert A, Boyko EJ, Iwashyna TJ, Maciejewski ML, Smith VA, Berkowitz TSZ, O'Hare AM, Viglianti EM, Aslan M, Bajema KL. Effectiveness of the 2023-to-2024 XBB.1.5 COVID-19 Vaccines Over Long-Term Follow-up : A Target Trial Emulation. Ann Intern Med 2025; 178:348-359. [PMID: 39903865 DOI: 10.7326/annals-24-01015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Monovalent COVID-19 vaccines targeting the XBB.1.5 Omicron variant were introduced in September 2023. In the absence of randomized controlled trials demonstrating their efficacy, information on real-world vaccine effectiveness (VE) is needed. OBJECTIVE To determine XBB.1.5 COVID-19 VE and the extent to which it declines over time. DESIGN Target trial emulation. SETTING U.S. Veterans Health Administration. PARTICIPANTS Eligible XBB.1.5 vaccine recipients were matched 1:1 to unvaccinated persons in 7 sequential biweekly trials with enrollment from 2 October 2023 through 3 January 2024. INTERVENTION XBB.1.5 COVID-19 vaccination versus no XBB.1.5 vaccination. MEASUREMENTS Outcomes were ascertained through 10 May 2024 and included any positive result on a SARS-CoV-2 test from day 10 after the matched index date, subsequent hospitalization within 1 day before or 10 days after the positive result, or death within 30 days after the positive result. Vaccine effectiveness was estimated as 100 × (1 - risk ratio). RESULTS Participants (91.3% male; mean age, 69.9 years) included 587 137 pairs of vaccinated and matched unvaccinated persons. Over a mean follow-up of 176 days (range, 118 to 211 days), VE was -3.26% (95% CI, -6.78% to -0.22%) against documented SARS-CoV-2 infection, 16.64% (CI, 6.47% to 25.77%) against SARS-CoV-2-associated hospitalization, and 26.61% (CI, 5.53% to 42.32%) against SARS-CoV-2-associated death. When estimated at 60, 90, and 120 days, respectively, VE against documented infection (14.21%, 7.29%, and 3.15%), hospitalization (37.57%, 30.84%, and 25.25%), or death (54.24%, 44.33%, and 30.25%) showed substantial waning. LIMITATION Potential for residual confounding and incomplete capture of COVID-19 vaccination and SARS-CoV-2-related outcomes. CONCLUSION COVID-19 vaccines targeting the XBB.1.5 variant of Omicron were not effective in preventing infection and had relatively low VE against hospitalization and death, which declined rapidly over time. PRIMARY FUNDING SOURCE U.S. Department of Veterans Affairs.
Collapse
Affiliation(s)
- George N Ioannou
- Research and Development and Division of Gastroenterology, Veterans Affairs Puget Sound Health Care System, and Division of Gastroenterology, University of Washington, Seattle, Washington (G.N.I.)
| | - Kristin Berry
- Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, Washington (K.B.)
| | - Nallakkandi Rajeevan
- Veterans Affairs Cooperative Studies Program Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, and Yale Center for Medical Informatics, Yale School of Medicine, New Haven, Connecticut (N.R.)
| | - Yuli Li
- Veterans Affairs Cooperative Studies Program Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut (Y.L.)
| | - Lei Yan
- Veterans Affairs Cooperative Studies Program Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, and Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut (L.Y., Y.H., H.-M.L.)
| | - Yuan Huang
- Veterans Affairs Cooperative Studies Program Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, and Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut (L.Y., Y.H., H.-M.L.)
| | - Hung-Mo Lin
- Veterans Affairs Cooperative Studies Program Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, and Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut (L.Y., Y.H., H.-M.L.)
| | - David Bui
- Veterans Affairs Portland Health Care System, Portland, Oregon (D.B.)
| | - Denise M Hynes
- Center of Innovation to Improve Veteran Involvement in Care, Veterans Affairs Portland Health Care System, Portland, and Health Management and Policy, College of Health, and Health Data and Informatics Program, Center for Quantitative Life Sciences, Oregon State University, Corvallis, Oregon (D.M.H.)
| | - Mazhgan Rowneki
- Center of Innovation to Improve Veteran Involvement in Care, Veterans Affairs Portland Health Care System, Portland, Oregon (M.R.)
| | - Amy Bohnert
- Center for Clinical Management Research, Veterans Affairs Ann Arbor Healthcare System, and Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan (A.B.)
| | - Edward J Boyko
- Seattle Epidemiologic Research and Information Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington (E.J.B.)
| | - Theodore J Iwashyna
- Center for Clinical Management Research, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, and Johns Hopkins University School of Medicine and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (T.J.I.)
| | - Matthew L Maciejewski
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Medical Center; Department of Population Health Sciences, Duke University School of Medicine; and Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina (M.L.M.)
| | - Valerie A Smith
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Medical Center; Department of Population Health Sciences, Duke University School of Medicine; Department of Biostatistics and Bioinformatics, Duke University School of Medicine; and Division of General Internal Medicine, Duke University School of Medicine, Durham, North Carolina (V.A.S.)
| | - Theodore S Z Berkowitz
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Medical Center, Durham, North Carolina (T.S.Z.B.)
| | - Ann M O'Hare
- Health Services Research and Development Center of Innovation for Veteran-Centered and Value-Driven Care, Veterans Affairs Puget Sound Health Care System, and Division of Nephrology, University of Washington, Seattle, Washington (A.M.O.)
| | - Elizabeth M Viglianti
- Center for Clinical Management Research, Veterans Affairs Ann Arbor Healthcare System, and Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan (E.M.V.)
| | - Mihaela Aslan
- Veterans Affairs Cooperative Studies Program Clinical Epidemiology Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, and Department of Medicine, Yale School of Medicine, New Haven, Connecticut (M.A.)
| | - Kristina L Bajema
- Veterans Affairs Portland Health Care System and Division of Infectious Diseases, Department of Medicine, Oregon Health & Science University, Portland, Oregon (K.L.B.)
| |
Collapse
|
14
|
Wrynla XH, Bates TA, Trank-Greene M, Wahedi M, Hinchliff A, Curlin ME, Tafesse FG. Immune imprinting and vaccination interval underly XBB.1.5 monovalent vaccine immunogenicity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.09.25321965. [PMID: 40034787 PMCID: PMC11875258 DOI: 10.1101/2025.02.09.25321965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
As COVID-19 transitions into endemicity and vaccines are annually updated to circulating SARS-CoV-2 lineages such as JN.1, exposure intervals and immune imprinting become critical considerations for vaccination strategy. Imprinting by the ancestral spike protein has been observed with the bivalent Wuhan-Hu-1/BA.4-5 vaccine and its persistence can be further evaluated in the context of the more recent XBB.1.5 monovalent vaccine. We assessed antibody responses in individuals who received three to four doses of Wuhan-Hu-1, one dose of bivalent Wuhan-Hu-1/BA.4-5, and one dose of XBB.1.5 vaccine (bivalent recipients). We compared these to individuals who received three to four doses of Wuhan-Hu-1 and one dose of XBB.1.5 vaccine without prior bivalent vaccination (bivalent non-recipients). Before XBB.1.5 vaccination, bivalent non-recipients demonstrated decreased breadth and potency of neutralizing antibodies compared to recipients, but at post-vaccination exhibited greater boosting of neutralizing antibodies against XBB.1.5 (18.4X versus 6.2X), EG.5.1 (30.9X versus 7.0X), and JN.1 (9.2X versus 3.7X) variants with trends toward higher neutralizing titers and comparable variant cross-neutralization. Increased boosting in non-recipients were similarly observed for IgA and total IgG/A/M isotypes binding the spike receptor-binding domain but not IgG nor IgM. Bivalent non-recipients had longer intervals between exposures, which has been reported to enhance antibody boosting; however, bivalent receipt and interval were tightly linked variables, preventing the isolation of individual contributions to boosting. Nonetheless, significant "back-boosting" of ancestral SARS-CoV-2 titers upon XBB.1.5 vaccination in both participant groups indicate that immune imprinting continues to affect contemporary vaccines. Altogether, our findings highlight imprinting and exposure intervals as important phenomena underlying variant-adapted COVID-19 vaccine immunogenicity.
Collapse
|
15
|
Lv H, Teo QW, Lee CCD, Liang W, Choi D, Mao KJ, Ardagh MR, Gopal AB, Mehta A, Szlembarski M, Bruzzone R, Wilson IA, Wu NC, Mok CKP. Differential antigenic imprinting effects between influenza H1N1 hemagglutinin and neuraminidase in a mouse model. J Virol 2025; 99:e0169524. [PMID: 39636110 PMCID: PMC11784018 DOI: 10.1128/jvi.01695-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/10/2024] [Indexed: 12/07/2024] Open
Abstract
Understanding how immune history influences influenza immunity is essential for developing effective vaccines and therapeutic strategies. This study examines the antigenic imprinting of influenza hemagglutinin (HA) and neuraminidase (NA) using a mouse model with sequential infections by H1N1 virus strains exhibiting substantial antigenic differences in HA. In our pre-2009 influenza infection model, we observed that mice with more extensive infection histories produced higher levels of functional NA-inhibiting antibodies (NAI). However, following further infection with the 2009 pandemic H1N1 strain, these mice demonstrated a reduced NAI to the challenged virus. Interestingly, prior exposure to older strains resulted in a lower HA antibody response (neutralization and HAI) to the challenged virus in both pre- and post-2009 scenarios, potentially due to faster viral clearance facilitated by immune memory recall. Overall, our findings reveal distinct trajectories in HA and NA immune responses, suggesting that immune imprinting can differentially impact these proteins based on the extent of antigenic variation in influenza viruses. IMPORTANCE Influenza viruses continue to pose a significant threat to human health, with vaccine effectiveness remaining a persistent challenge. Individual immune history is a crucial factor that can influence antibody responses to subsequent influenza exposures. While many studies have explored how pre-existing antibodies shape the induction of anti-HA antibodies following influenza virus infections or vaccinations, the impact on anti-NA antibodies has been less extensively studied. Using a mouse model, our study demonstrates that within pre-2009 H1N1 strains, an extensive immune history negatively impacted anti-HA antibody responses but enhanced anti-NA antibody responses. However, in response to the 2009 pandemic H1N1 strain, which experienced an antigenic shift, both anti-HA and anti-NA antibody responses were hindered by antibodies from prior pre-2009 H1N1 virus infections. These findings provide important insights into how antigenic imprinting affects both anti-HA and anti-NA antibody responses and underscore the need to consider immune history in developing more effective influenza vaccination strategies.
Collapse
MESH Headings
- Animals
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Neuraminidase/immunology
- Neuraminidase/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Mice
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Disease Models, Animal
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Female
- Humans
- Mice, Inbred BALB C
- Influenza Vaccines/immunology
- Antigens, Viral/immunology
- Antibodies, Neutralizing/immunology
- Influenza, Human/immunology
- Influenza, Human/virology
- Antigenic Variation
Collapse
Affiliation(s)
- Huibin Lv
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qi Wen Teo
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chang-Chun D. Lee
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, California, USA
| | - Weiwen Liang
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Centre for Immunology & Infection, Hong Kong Science Park, Hong Kong, China
| | - Danbi Choi
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Kevin J. Mao
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Madison R. Ardagh
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Akshita B. Gopal
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Arjun Mehta
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Matt Szlembarski
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Centre for Immunology & Infection, Hong Kong Science Park, Hong Kong, China
- Istituto Pasteur Italia, Rome, Italy
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, California, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Nicholas C. Wu
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Chris K. P. Mok
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- S.H. Ho Research Centre for Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Oluka GK, Sembera J, Katende JS, Ankunda V, Kato L, Kurshan A, Graham C, Seow J, Doores KJ, Malim MH, Fox JM, Kaleebu P, Serwanga J. Long-Term Immune Consequences of Initial SARS-CoV-2 A.23.1 Exposure: A Longitudinal Study of Antibody Responses and Cross-Neutralization in a Ugandan Cohort. Vaccines (Basel) 2025; 13:143. [PMID: 40006690 PMCID: PMC11860332 DOI: 10.3390/vaccines13020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Background: This study assessed the long-term dynamics of neutralizing antibodies in a Ugandan cohort primarily exposed to the A.23.1 SARS-CoV-2 variant, examining how this shaped immune breadth and potency against diverse strains following infection and prototype-based vaccination. Methods: We conducted a 427-day retrospective analysis of 41 participants across multiple SARS-CoV-2 waves, assessing binding and neutralizing antibody responses using in-house ELISA and pseudotyped virus neutralization assays. We quantified immune responses to key SARS-CoV-2 variants, A.23.1, D614G, Delta, and BA.4, capturing evolving immunity across the pandemic. Results: Neutralizing antibody titers against A.23.1 remained significantly higher than those against D614G, Delta, and BA.4, highlighting the solid immune memory following A.23.1 infection. Consistently lower titers were observed for BA.4 across all time points, aligning with its strong immune-evasion capability. Correlations between neutralizing titers and spike-directed IgG (S-IgG) concentrations were significantly stronger for A.23.1 than for D614G, with no correlation for BA.4. ChAdOx1-S vaccination substantially elevated the neutralizing titers across all variants, most notably BA.4, highlighting the essential role of vaccination in boosting immunity, even in individuals with initially low titers. Conclusions: Initial exposure to the A.23.1 variant triggered potent immune responses, shaping neutralizing antibody dynamics during subsequent exposures. These findings highlight the importance of accounting for early viral exposures in vaccine development and public health planning. The distinctly lower immune response to BA.4 highlights the need for continuous antigenic monitoring and timely vaccine updates for protection against emerging variants. Vaccination remains essential for reinforcing and sustaining immunity against evolving variants.
Collapse
Affiliation(s)
- Gerald Kevin Oluka
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine (MRC/UVRI & LSHTM) Research Unit, Entebbe 26, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe 26, Uganda
| | - Jackson Sembera
- Department of Immunology, Uganda Virus Research Institute, Entebbe 26, Uganda
| | - Joseph Ssebwana Katende
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine (MRC/UVRI & LSHTM) Research Unit, Entebbe 26, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe 26, Uganda
| | - Violet Ankunda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine (MRC/UVRI & LSHTM) Research Unit, Entebbe 26, Uganda
| | - Laban Kato
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine (MRC/UVRI & LSHTM) Research Unit, Entebbe 26, Uganda
| | - Ashwini Kurshan
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Carl Graham
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Jeffrey Seow
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Katie J. Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Michael H. Malim
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Julie M. Fox
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King’s College London, London SE1 9RT, UK
| | - Pontiano Kaleebu
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine (MRC/UVRI & LSHTM) Research Unit, Entebbe 26, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe 26, Uganda
| | - Jennifer Serwanga
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine (MRC/UVRI & LSHTM) Research Unit, Entebbe 26, Uganda
- Department of Immunology, Uganda Virus Research Institute, Entebbe 26, Uganda
| |
Collapse
|
17
|
Rössler A, Netzl A, Lasrado N, Chaudhari J, Mühlemann B, Wilks SH, Kimpel J, Smith DJ, Barouch DH. Nonhuman primate antigenic cartography of SARS-CoV-2. Cell Rep 2025; 44:115140. [PMID: 39754717 PMCID: PMC11781863 DOI: 10.1016/j.celrep.2024.115140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/06/2025] Open
Abstract
Virus neutralization profiles against primary infection sera and corresponding antigenic cartography are integral part of the COVID-19 and influenza vaccine strain selection processes. Human single variant exposure sera have previously defined the antigenic relationships among SARS-CoV-2 variants but are now largely unavailable due to widespread population immunity. Therefore, antigenic characterization of future SARS-CoV-2 variants will require an animal model, analogous to using ferrets for influenza virus. We evaluated neutralization profiles against 23 SARS-CoV-2 variants in nonhuman primates (NHPs) after single variant exposure and generated an NHP-derived antigenic map. We identified a distant antigenic region occupied by BA.2.86, JN.1, and the descendants KP.2, KP.3, and KZ.1.1.1. We also found that the monovalent XBB.1.5 mRNA vaccine induced broad immunity against the mapped antigenic space. In addition, substantial concordance was observed between our NHP-derived and two human antigenic maps, demonstrating the utility of NHPs as a surrogate for antigenic cartography in humans.
Collapse
Affiliation(s)
- Annika Rössler
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Antonia Netzl
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, CB2 3EJ, Cambridge, Cambridgeshire, UK
| | - Ninaad Lasrado
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jayeshbhai Chaudhari
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Barbara Mühlemann
- Institute of Virology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Berlin, Germany; German Centre for Infection Research (DZIF), Partner Site Charité, 10117 Berlin, Berlin, Germany
| | - Samuel H Wilks
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, CB2 3EJ, Cambridge, Cambridgeshire, UK
| | - Janine Kimpel
- Institute of Virology, Department of Hygiene, Microbiology and Virology, Medical University of Innsbruck, Innsbruck, Tyrol 6020, Austria
| | - Derek J Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, CB2 3EJ, Cambridge, Cambridgeshire, UK
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
18
|
Dadonaite B, Burrell AR, Logue J, Chu HY, Payne DC, Haslam DB, Staat MA, Bloom JD. SARS-CoV-2 neutralizing antibody specificities differ dramatically between recently infected infants and immune-imprinted individuals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633612. [PMID: 39896663 PMCID: PMC11785066 DOI: 10.1101/2025.01.17.633612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The immune response to viral infection is shaped by past exposures to related virus strains, a phenomenon known as imprinting. For SARS-CoV-2, much of the population has been imprinted by a viral spike from an early strain, either through vaccination or infection during the early stages of the COVID-19 pandemic. As a consequence of this imprinting, infection with more recent SARS-CoV-2 strains primarily boosts cross-reactive antibodies elicited by the imprinting strain. Here we compare the neutralizing antibody specificities of imprinted individuals versus infants infected with a recent strain. Specifically, we use pseudovirus-based deep mutational scanning to measure how spike mutations affect neutralization by the serum antibodies of adults and children imprinted by the original vaccine versus infants with a primary infection by a XBB* variant. While the serum neutralizing activity of the imprinted individuals primarily targets the spike receptor-binding domain (RBD), serum neutralizing activity of infants only infected with XBB* mostly targets the spike N-terminal domain (NTD). In these infants, secondary exposure to the XBB* spike via vaccination shifts more of the neutralizing activity towards the RBD, although the specific RBD sites targeted are different than for imprinted adults. The dramatic differences in neutralization specificities among individuals with different exposure histories likely impact SARS-CoV-2 evolution.
Collapse
Affiliation(s)
- Bernadeta Dadonaite
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
| | - Allison R. Burrell
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jenni Logue
- University of Washington, Department of Medicine, Division of Allergy and Infectious Diseases, Seattle, WA
| | - Helen Y. Chu
- University of Washington, Department of Medicine, Division of Allergy and Infectious Diseases, Seattle, WA
| | - Daniel C. Payne
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - David B. Haslam
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mary A. Staat
- Department of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, 98109, USA
- Howard Hughes Medical Institute, Seattle, WA, 98195, USA
| |
Collapse
|
19
|
Wang Z, Li L, Du R, Chen X, Sun Y, Qin R, Li Y, Feng H, Hu L, Chen X, Lu M, Jiang L, Zuo T. Ancestral SARS-CoV-2 immune imprinting persists on RBD but not NTD after sequential Omicron infections. iScience 2025; 28:111557. [PMID: 39807166 PMCID: PMC11728909 DOI: 10.1016/j.isci.2024.111557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/08/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025] Open
Abstract
Whether Omicron exposures could overcome ancestral SARS-CoV-2 immune imprinting remains controversial. Here we analyzed B cell responses evoked by sequential Omicron infections in vaccinated and unvaccinated individuals. Plasma neutralizing antibody titers against ancestral SARS-CoV-2 and variants indicate that immune imprinting is not consistently induced by inactivated or recombinant protein vaccines. However, once robustly induced, immune imprinting is not countered by successive Omicron challenges. We compared binding specificities, neutralizing capacities, developing origins and targeting epitopes of monoclonal antibodies from those individuals. Although receptor-binding domain (RBD) and N-terminal domain (NTD) of spike are both primary targets for neutralizing antibodies, immune imprinting only shapes antibody responses to RBD by impeding the production of Omicron-specific neutralizing antibodies while facilitating the development of broadly neutralizing antibodies. We propose that immune imprinting can be either neglected by NTD-based vaccines to induce variant-specific antibodies or leveraged by RBD-containing vaccines to induce broadly neutralizing antibodies.
Collapse
Affiliation(s)
- Zuowei Wang
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Ling Li
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Ruiping Du
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xixian Chen
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yi Sun
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Rongrong Qin
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yunjian Li
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Hualong Feng
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Lin Hu
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xuanyi Chen
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Maosheng Lu
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Liwei Jiang
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Teng Zuo
- Laboratory of Immunoengineering, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
20
|
Jian F, Wang J, Yisimayi A, Song W, Xu Y, Chen X, Niu X, Yang S, Yu Y, Wang P, Sun H, Yu L, Wang J, Wang Y, An R, Wang W, Ma M, Xiao T, Gu Q, Shao F, Wang Y, Shen Z, Jin R, Cao Y. Evolving antibody response to SARS-CoV-2 antigenic shift from XBB to JN.1. Nature 2025; 637:921-929. [PMID: 39510125 PMCID: PMC11754117 DOI: 10.1038/s41586-024-08315-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
The continuous evolution of SARS-CoV-2, particularly the emergence of the BA.2.86/JN.1 lineage replacing XBB, necessitates re-evaluation of vaccine compositions1-3. Here, we provide a comprehensive analysis of the humoral immune response to XBB and JN.1 human exposure. We demonstrate the antigenic distinctiveness of XBB and JN.1 lineages in SARS-CoV-2-naive individuals and show that infection with JN.1 elicits superior plasma neutralization against its subvariants. We highlight the strong immune evasion and receptor-binding capability of KP.3, supporting its foreseeable prevalence. Extensive analysis of the B cell receptor repertoire, in which we isolate approximately 2,000 receptor-binding-domain-specific antibodies, with targeting epitopes characterized by deep mutational scanning, underscores the superiority of JN.1-elicited memory B cells4,5. Class 1 IGHV3-53/3-66-derived neutralizing antibodies (NAbs) are important contributors to the wild-type reactivity of NAbs against JN.1. However, KP.2 and KP.3 evade a substantial subset of these antibodies, even those induced by JN.1, supporting a need for booster updates. JN.1-induced Omicron-specific antibodies also demonstrate high potency across Omicron. Escape hotspots for these NAbs have already been mutated, resulting in a higher immune barrier to escape and indicating probable recovery of escaped NAbs. In addition, the prevalence of IGHV3-53/3-66-derived antibodies and their ability to compete with all Omicron-specific NAbs suggests that they have an inhibitory effect on the activation of Omicron-specific naive B cells, potentially explaining the heavy immune imprinting in mRNA-vaccinated individuals6-8. These findings delineate the evolving antibody response to the antigenic shift of Omicron from XBB to JN.1 and highlight the importance of developing the JN.1 lineage, especially KP.2- and KP.3-based vaccine boosters.
Collapse
Affiliation(s)
- Fanchong Jian
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- Changping Laboratory, Beijing, China
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Jing Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- Changping Laboratory, Beijing, China
- School of Life Sciences, Peking University, Beijing, China
| | - Ayijiang Yisimayi
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- Changping Laboratory, Beijing, China
- School of Life Sciences, Peking University, Beijing, China
| | - Weiliang Song
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- Changping Laboratory, Beijing, China
- School of Life Sciences, Peking University, Beijing, China
| | - Yanli Xu
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaosu Chen
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiao Niu
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Sijie Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
| | | | - Peng Wang
- Changping Laboratory, Beijing, China
| | | | | | - Jing Wang
- Changping Laboratory, Beijing, China
| | - Yao Wang
- Changping Laboratory, Beijing, China
| | - Ran An
- Changping Laboratory, Beijing, China
| | | | | | - Tianhe Xiao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-NIBS, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | | - Fei Shao
- Changping Laboratory, Beijing, China
| | - Youchun Wang
- Changping Laboratory, Beijing, China
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Zhongyang Shen
- Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Ronghua Jin
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yunlong Cao
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China.
- Changping Laboratory, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
21
|
Maltseva M, Keeshan A, Cooper C, Langlois MA. Immune imprinting: The persisting influence of the first antigenic encounter with rapidly evolving viruses. Hum Vaccin Immunother 2024; 20:2384192. [PMID: 39149872 PMCID: PMC11328881 DOI: 10.1080/21645515.2024.2384192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/17/2024] Open
Abstract
Immune imprinting is a phenomenon that stems from the fundamentals of immunological memory. Upon recurrent exposures to an evolving pathogen, the immune system must weigh the benefits of rapidly recalling established antibody repertoires with greater affinity to the initial variant or invest additional time and energy in producing de novo responses specific to the emerging variant. In this review, we delve into the mechanistic complexities of immune imprinting and its role in shaping subsequent immune responses, both de novo and recall, against rapidly evolving respiratory viruses such as influenza and coronaviruses. By exploring the duality of immune imprinting, we examine its potential to both enhance or hinder immune protection against disease, while emphasizing the role of host and viral factors. Finally, we explore how different vaccine platforms may affect immune imprinting and comment on vaccine strategies that can favor de novo variant-specific antibody responses.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alexa Keeshan
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Curtis Cooper
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, Ottawa Hospital Research Institute Clinical Epidemiology Program, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
22
|
Brangel P, Tureli S, Mühlemann B, Liechti N, Zysset D, Engler O, Hunger-Glaser I, Ghiga I, Mattiuzzo G, Eckerle I, Bekliz M, Rössler A, Schmitt MM, Knabl L, Kimpel J, Tort LFL, de Araujo MF, de Oliveira ACA, Caetano BC, Siqueira MM, Budt M, Gensch JM, Wolff T, Hassan T, Selvaraj FA, Hermanus T, Kgagudi P, Crowther C, Richardson SI, Bhiman JN, Moore PL, Cheng SMS, Li JKC, Poon LLM, Peiris M, Corman VM, Drosten C, Lai L, Hunsawong T, Rungrojcharoenkit K, Lohachanakul J, Sigal A, Khan K, Thiel V, Barut GT, Ebert N, Mykytyn AZ, Owusu Donkor I, Aboagye JO, Nartey PA, Van Kerkhove MD, Cunningham J, Haagmans BL, Suthar MS, Smith D, Subissi L. A Global Collaborative Comparison of SARS-CoV-2 Antigenicity Across 15 Laboratories. Viruses 2024; 16:1936. [PMID: 39772242 PMCID: PMC11680265 DOI: 10.3390/v16121936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Setting up a global SARS-CoV-2 surveillance system requires an understanding of how virus isolation and propagation practices, use of animal or human sera, and different neutralisation assay platforms influence assessment of SARS-CoV-2 antigenicity. In this study, with the contribution of 15 independent laboratories across all WHO regions, we carried out a controlled analysis of neutralisation assay platforms using the first WHO International Standard for antibodies to SARS-CoV-2 variants of concern (source: NIBSC). Live virus isolates (source: WHO BioHub or individual labs) or spike plasmids (individual labs) for pseudovirus production were used to perform neutralisation assays using the same serum panels. When comparing fold drops, excellent data consistency was observed across the labs using common reagents, including between pseudovirus and live virus neutralisation assays (RMSD of data from mean fold drop was 0.59). Utilising a Bayesian model, geometric mean titres and assay titre magnitudes (offsets) can describe the data efficiently. Titre magnitudes were seen to vary largely even for labs within the same assay group. We have observed that overall, live Microneutralisation assays tend to have the lowest titres, whereas Pseudovirus Neutralisation have the highest (with a mean difference of 3.2 log2 units between the two). These findings are relevant for laboratory networks, such as the WHO Coronavirus Laboratory Network (CoViNet), that seek to support a global surveillance system for evolution and antigenic characterisation of variants to support monitoring of population immunity and vaccine composition policy.
Collapse
Affiliation(s)
| | - Sina Tureli
- Centre for Pathogen Evolution, University of Cambridge, Cambridge CB3 0FD, UK
| | - Barbara Mühlemann
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Virology, Charitéplatz 1, 10117 Berlin, Germany
- German Centre for Infection Research (DZIF), Associated Partner Site Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicole Liechti
- WHO BioHub Facility, Spiez Laboratory, 3700 Spiez, Switzerland
| | - Daniel Zysset
- WHO BioHub Facility, Spiez Laboratory, 3700 Spiez, Switzerland
| | - Olivier Engler
- WHO BioHub Facility, Spiez Laboratory, 3700 Spiez, Switzerland
| | | | - Ioana Ghiga
- World Health Organization, 1202 Geneva, Switzerland
| | - Giada Mattiuzzo
- Medicines and Healthcare Products Regulatory Agency, London SW1W 9SZ, UK
| | - Isabella Eckerle
- Department of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Division of Infectious Diseases, Geneva University Hospitals, 1205 Geneva, Switzerland
- Geneva Centre for Emerging Viral Diseases, University Hospitals of Geneva and University of Geneva, 1205 Geneva, Switzerland
| | - Meriem Bekliz
- Department of Medicine, University of Geneva, 1205 Geneva, Switzerland
- Geneva Centre for Emerging Viral Diseases, University Hospitals of Geneva and University of Geneva, 1205 Geneva, Switzerland
| | - Annika Rössler
- Institute of Virology, Department of Hygiene, Microbiology and Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Melanie M. Schmitt
- Institute of Virology, Department of Hygiene, Microbiology and Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Ludwig Knabl
- Tyrolpath Obrist Brunhuber GmbH, 6511 Zams, Austria
| | - Janine Kimpel
- Institute of Virology, Department of Hygiene, Microbiology and Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Luis Fernando Lopez Tort
- Laboratory of Respiratory, Exanthematous and Enteric Viruses and Viral Emergencies (LVRE), Oswaldo Cruz Institute (IOC-Fiocruz), Rio de Janeiro 21.040-900, Brazil
- Laboratory of Molecular Virology, Department of Biological Sciences, CENUR Litoral Norte, Universidad de la República, Salto 50000, Uruguay
| | - Mia Ferreira de Araujo
- Laboratory of Respiratory, Exanthematous and Enteric Viruses and Viral Emergencies (LVRE), Oswaldo Cruz Institute (IOC-Fiocruz), Rio de Janeiro 21.040-900, Brazil
| | - Any Caroline Alves de Oliveira
- Laboratory of Respiratory, Exanthematous and Enteric Viruses and Viral Emergencies (LVRE), Oswaldo Cruz Institute (IOC-Fiocruz), Rio de Janeiro 21.040-900, Brazil
| | - Braulia Costa Caetano
- Laboratory of Respiratory, Exanthematous and Enteric Viruses and Viral Emergencies (LVRE), Oswaldo Cruz Institute (IOC-Fiocruz), Rio de Janeiro 21.040-900, Brazil
| | - Marilda Mendonça Siqueira
- Laboratory of Respiratory, Exanthematous and Enteric Viruses and Viral Emergencies (LVRE), Oswaldo Cruz Institute (IOC-Fiocruz), Rio de Janeiro 21.040-900, Brazil
| | - Matthias Budt
- Unit 17 “Influenza and Other Respiratory Viruses”, Robert Koch Institut, 13353 Berlin, Germany
| | - Jean-Marc Gensch
- Unit 17 “Influenza and Other Respiratory Viruses”, Robert Koch Institut, 13353 Berlin, Germany
| | - Thorsten Wolff
- Unit 17 “Influenza and Other Respiratory Viruses”, Robert Koch Institut, 13353 Berlin, Germany
| | - Tarteel Hassan
- Reference Laboratory for Infectious Diseases, Purelab, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates
| | - Francis Amirtharaj Selvaraj
- Reference Laboratory for Infectious Diseases, Purelab, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates
| | - Tandile Hermanus
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2001, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2131, South Africa
| | - Prudence Kgagudi
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2001, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2131, South Africa
| | - Carol Crowther
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2001, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2131, South Africa
| | - Simone I. Richardson
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2001, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2131, South Africa
| | - Jinal N. Bhiman
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2001, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2131, South Africa
| | - Penny L. Moore
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2001, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2131, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban 4001, South Africa
| | - Samuel M. S. Cheng
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - John K. C. Li
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Leo L. M. Poon
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Malik Peiris
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Victor M. Corman
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Virology, Charitéplatz 1, 10117 Berlin, Germany
- German Centre for Infection Research (DZIF), Associated Partner Site Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Christian Drosten
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Virology, Charitéplatz 1, 10117 Berlin, Germany
- German Centre for Infection Research (DZIF), Associated Partner Site Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Lilin Lai
- Department of Pediatrics, Center for Childhood Infections and Vaccines, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Taweewun Hunsawong
- Walter Reed Army Institute of Research-Armed Forces Research Institute of Medical Sciences, Bangkok 10500, Thailand
| | - Kamonthip Rungrojcharoenkit
- Walter Reed Army Institute of Research-Armed Forces Research Institute of Medical Sciences, Bangkok 10500, Thailand
| | - Jindarat Lohachanakul
- Walter Reed Army Institute of Research-Armed Forces Research Institute of Medical Sciences, Bangkok 10500, Thailand
| | - Alex Sigal
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Africa Health Research Institute, Durban 4013, South Africa
| | - Khadija Khan
- Africa Health Research Institute, Durban 4013, South Africa
| | - Volker Thiel
- Multidisciplinary Center for Infectious Diseases, University of Bern, 3012 Bern, Switzerland
- Institute of Virology and Immunology, Mittelhäusern and Bern, 3012 Bern, Switzerland
| | - G. Tuba Barut
- Multidisciplinary Center for Infectious Diseases, University of Bern, 3012 Bern, Switzerland
- Institute of Virology and Immunology, Mittelhäusern and Bern, 3012 Bern, Switzerland
| | - Nadine Ebert
- Multidisciplinary Center for Infectious Diseases, University of Bern, 3012 Bern, Switzerland
- Institute of Virology and Immunology, Mittelhäusern and Bern, 3012 Bern, Switzerland
| | | | - Irene Owusu Donkor
- Medical and Scientific Research Centre, University of Ghana Medical Centre, Accra P.O. Box LG 25, Ghana
| | - James Odame Aboagye
- Medical and Scientific Research Centre, University of Ghana Medical Centre, Accra P.O. Box LG 25, Ghana
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon P.O. Box LG 581, Ghana
| | - Prince Adom Nartey
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon P.O. Box LG 581, Ghana
| | | | | | | | - Mehul S. Suthar
- Department of Pediatrics, Center for Childhood Infections and Vaccines, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Derek Smith
- Centre for Pathogen Evolution, University of Cambridge, Cambridge CB3 0FD, UK
| | | |
Collapse
|
23
|
Chu HY. A Perspective on Vaccinology. J Infect Dis 2024; 230:1302-1304. [PMID: 39400196 DOI: 10.1093/infdis/jiae506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- Helen Y Chu
- Departments of Medicine and Epidemiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
24
|
Rosen LE, Tortorici MA, De Marco A, Pinto D, Foreman WB, Taylor AL, Park YJ, Bohan D, Rietz T, Errico JM, Hauser K, Dang HV, Chartron JW, Giurdanella M, Cusumano G, Saliba C, Zatta F, Sprouse KR, Addetia A, Zepeda SK, Brown J, Lee J, Dellota E, Rajesh A, Noack J, Tao Q, DaCosta Y, Tsu B, Acosta R, Subramanian S, de Melo GD, Kergoat L, Zhang I, Liu Z, Guarino B, Schmid MA, Schnell G, Miller JL, Lempp FA, Czudnochowski N, Cameroni E, Whelan SPJ, Bourhy H, Purcell LA, Benigni F, di Iulio J, Pizzuto MS, Lanzavecchia A, Telenti A, Snell G, Corti D, Veesler D, Starr TN. A potent pan-sarbecovirus neutralizing antibody resilient to epitope diversification. Cell 2024; 187:7196-7213.e26. [PMID: 39383863 PMCID: PMC11645210 DOI: 10.1016/j.cell.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolution has resulted in viral escape from clinically authorized monoclonal antibodies (mAbs), creating a need for mAbs that are resilient to epitope diversification. Broadly neutralizing coronavirus mAbs that are sufficiently potent for clinical development and retain activity despite viral evolution remain elusive. We identified a human mAb, designated VIR-7229, which targets the viral receptor-binding motif (RBM) with unprecedented cross-reactivity to all sarbecovirus clades, including non-ACE2-utilizing bat sarbecoviruses, while potently neutralizing SARS-CoV-2 variants since 2019, including the recent EG.5, BA.2.86, and JN.1. VIR-7229 tolerates extraordinary epitope variability, partly attributed to its high binding affinity, receptor molecular mimicry, and interactions with RBM backbone atoms. Consequently, VIR-7229 features a high barrier for selection of escape mutants, which are rare and associated with reduced viral fitness, underscoring its potential to be resilient to future viral evolution. VIR-7229 is a strong candidate to become a next-generation medicine.
Collapse
MESH Headings
- Humans
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Epitopes/immunology
- Epitopes/chemistry
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/chemistry
- Antibodies, Viral/immunology
- Antibodies, Viral/chemistry
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/metabolism
- Spike Glycoprotein, Coronavirus/genetics
- Cross Reactions/immunology
- Chiroptera/virology
- Chiroptera/immunology
- COVID-19/immunology
- COVID-19/virology
- Angiotensin-Converting Enzyme 2/metabolism
- Angiotensin-Converting Enzyme 2/chemistry
Collapse
Affiliation(s)
| | | | - Anna De Marco
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Dora Pinto
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - William B Foreman
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ashley L Taylor
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Dana Bohan
- Vir Biotechnology, San Francisco, CA 94158, USA
| | - Tyson Rietz
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | | | - Ha V Dang
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | - Martina Giurdanella
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Giuseppe Cusumano
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Christian Saliba
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Fabrizia Zatta
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Kaitlin R Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Samantha K Zepeda
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jack Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jimin Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | | | - Julia Noack
- Vir Biotechnology, San Francisco, CA 94158, USA
| | - Qiqing Tao
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | - Brian Tsu
- Vir Biotechnology, San Francisco, CA 94158, USA
| | - Rima Acosta
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | - Guilherme Dias de Melo
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015 Paris, France
| | - Lauriane Kergoat
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015 Paris, France
| | - Ivy Zhang
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Barbara Guarino
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Michael A Schmid
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | | | - Florian A Lempp
- Vir Biotechnology, San Francisco, CA 94158, USA; Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | - Elisabetta Cameroni
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hervé Bourhy
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, F-75015 Paris, France
| | | | - Fabio Benigni
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | | | - Antonio Lanzavecchia
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | | | | | - Davide Corti
- Humabs BioMed SA, a Subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland.
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| | - Tyler N Starr
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
25
|
Malladi SK, Jaiswal D, Ying B, Alsoussi WB, Darling TL, Dadonaite B, Civljak A, Horvath SC, Zhou JQ, Kim W, Turner JS, Schmitz AJ, Han F, Scheaffer SM, Farnsworth CW, Nachbagauer R, Nestorova B, Chalkias S, Klebert MK, Edwards DK, Paris R, Strnad BS, Middleton WD, O’Halloran JA, Presti RM, Bloom JD, Boon ACM, Diamond MS, Bajic G, Ellebedy AH. Defining a highly conserved B cell epitope in the receptor binding motif of SARS-CoV-2 spike glycoprotein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.625234. [PMID: 39713327 PMCID: PMC11661108 DOI: 10.1101/2024.12.06.625234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
SARS-CoV-2 mRNA vaccines induce robust and persistent germinal centre (GC) B cell responses in humans. It remains unclear how the continuous evolution of the virus impacts the breadth of the induced GC B cell response. Using ultrasound-guided fine needle aspiration, we examined draining lymph nodes of nine healthy adults following bivalent booster immunization. We show that 77.8% of the B cell clones in the GC expressed as representative monoclonal antibodies recognized the spike protein, with a third (37.8%) of these targeting the receptor binding domain (RBD). Strikingly, only one RBD-targeting mAb, mAb-52, neutralized all tested SARS-CoV-2 strains, including the recent KP.2 variant. mAb-52 utilizes the IGHV3-66 public clonotype, protects hamsters challenged against the EG.5.1 variant and targets the class I/II RBD epitope, closely mimicking the binding footprint of ACE2. Finally, we show that the remarkable breadth of mAb-52 is due to the somatic hypermutations accumulated within vaccine-induced GC reaction.
Collapse
Affiliation(s)
- Sameer Kumar Malladi
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Deepika Jaiswal
- Department of Microbiology, Icahn School of Medicine at Mount Sinai; New York, NY, USA
| | - Baoling Ying
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Wafaa B. Alsoussi
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Tamarand L. Darling
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Bernadeta Dadonaite
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center; Seattle, WA, USA
| | - Alesandro Civljak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai; New York, NY, USA
| | - Stephen C. Horvath
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Julian Q. Zhou
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Department of Microbiology, Korea University College of Medicine; Seoul, Korea
| | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Aaron J. Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Fangjie Han
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Suzanne M. Scheaffer
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Christopher W. Farnsworth
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | | | | | | | - Michael K. Klebert
- Clinical Trials Unit, Washington University School of Medicine; St. Louis, MO, USA
| | | | | | - Benjamin S. Strnad
- Mallinckrodt Institute of Radiology, Washington University School of Medicine; St. Louis, MO, USA
| | - William D. Middleton
- Mallinckrodt Institute of Radiology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jane A. O’Halloran
- Division of Infectious Diseases, Washington University School of Medicine; St. Louis, MO, USA
| | - Rachel M. Presti
- Division of Infectious Diseases, Washington University School of Medicine; St. Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine; St. Louis, MO, USA
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center; Seattle, WA, USA
- Howard Hughes Medical Institute; Seattle, WA, USA
| | - Adrianus C. M. Boon
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine; St. Louis, MO, USA
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine; St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine; St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine; St. Louis, MO, USA
| | - Goran Bajic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai; New York, NY, USA
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine; St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine; St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine; St. Louis, MO, USA
| |
Collapse
|
26
|
Li Y, Qin S, Dong L, Xiao Y, Zhang Y, Hou Y, Qiao S, Zhang R, Li Y, Bao Y, Zhao X, Ma Y, Gao GF. Multi-omic characteristics of longitudinal immune profiling after breakthrough infections caused by Omicron BA.5 sublineages. EBioMedicine 2024; 110:105428. [PMID: 39536392 PMCID: PMC11605469 DOI: 10.1016/j.ebiom.2024.105428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Omicron sub-variants breakthrough infections (BTIs) have led to millions of coronavirus disease 2019 (COVID-19) cases worldwide. The acute-phase immune status is critical for prognosis, however, the dynamic immune profiling of COVID-19 during the first month after BTIs remains unclear. METHODS In this study, we monitored the immune dynamics at various timepoints in a longitudinal cohort during the first month post-BTIs through clinical evaluation, single-cell RNA sequencing (scRNA-seq), T cell receptor (TCR)/B cell receptor (BCR) sequencing, and antibody mass spectrometry. FINDINGS Serological analysis revealed limited impairment to functions of major organs, active cellular and humoral immunity at 2 weeks post-BTI, with significant increases in cytokines (CKs) and neutralizing antibody levels. However, 1 month post-BTI, organ function parameters and CK levels reverted to pre-infection levels, whereas neutralizing antibody levels remained high. Notably, scRNA-seq showed that lymphocytes maintained strong antiviral activity and cell depletion at 2 weeks and 1 month post-BTI, with genes CD81, ABHD17A, CXCR4, DUSP1, etc. upregulated, and genes PFDN5, DYNLRB1, CD52, etc. downregulated, indicating that lymphocytes status take longer to recover to normal levels than that routine blood tests revealed. Additionally, T cell-exhaustion associated genes, including LAG3, TIGIT, PDCD1, CTLA4, HAVCR2, and TOX, were upregulated after BTI. TCRs and BCRs exhibited higher clonotypes, mainly in CD8Tem or plasmablast cells, at 2 weeks post-BTI comparing 1 month. More IgG and IgA-type BCRs were found in the groups of 1 month post-BTI, with higher somatic hypermutation, indicating greater maturity. Verification of monoclonal antibodies corresponding to amplified BCRs highlighted the antigen-specific and broad-spectrum characteristics. INTERPRETATION Our study elucidated the dynamic immune profiling of individuals after Omicron BA.5 sublineages BTI. Strong immune activation, antiviral response, antibody maturation and class transition at 2 weeks and 1 month after BTI may provide essential insights into pathogenicity, sequential immune status, recovery mechanisms of Omicron sublineage BTI. FUNDING This study was supported by the National Key R&D Program of China, the China Postdoctoral Science Foundation, Guangdong Basic and Applied Basic Research Foundation, the National Natural Science Foundation of China, CAS Project for Young Scientists in Basic Research, and the Air Force Special Medical Center Science and Technology Booster Program.
Collapse
MESH Headings
- Humans
- COVID-19/immunology
- COVID-19/virology
- SARS-CoV-2/immunology
- Male
- Female
- Middle Aged
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Longitudinal Studies
- Adult
- Cytokines/metabolism
- Aged
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Single-Cell Analysis
- Breakthrough Infections
- Multiomics
Collapse
Affiliation(s)
- Yanhua Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shijie Qin
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; Shenzhen Children's Hospital, Shenzhen, 518026, China; The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Lei Dong
- Department of Clinical Laboratory, Air Force Medical Center, Beijing, 100142, China
| | - Yunfeng Xiao
- Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yanan Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; Shenzhen Children's Hospital, Shenzhen, 518026, China
| | - Yali Hou
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shitong Qiao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Rong Zhang
- Guangxi University State Key Laboratory for Conservation and Utilization of Subtropical Agro BioResources, Nanning, 53000, China
| | - Ying Li
- Department of Clinical Laboratory, Air Force Medical Center, Beijing, 100142, China
| | - Yanmin Bao
- Shenzhen Children's Hospital, Shenzhen, 518026, China.
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; Beijing Life Science Academy, Beijing, 102209, China.
| | - Yueyun Ma
- Department of Clinical Laboratory, Air Force Medical Center, Beijing, 100142, China.
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
27
|
Jia T, Wang F, Chen Y, Liao G, Xu Q, Chen J, Wu J, Li N, Wang L, Yuan L, Wang D, Xie Q, Luo C, Luo H, Wang Y, Chen Y, Shu Y. Expanded immune imprinting and neutralization spectrum by hybrid immunization following breakthrough infections with SARS-CoV-2 variants after three-dose vaccination. J Infect 2024; 89:106362. [PMID: 39608577 DOI: 10.1016/j.jinf.2024.106362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/28/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Despite vaccination, SARS-CoV-2 evolution leads to breakthrough infections and reinfections worldwide. Knowledge of hybrid immunization is crucial for future broad-spectrum SARS-CoV-2 vaccines. METHODS In this study, we investigated neutralizing antibodies (nAbs) against the SARS-CoV-2 ancestral virus (wild-type [WT]), pre-Omicron VOCs, Omicron subvariants, and SARS-CoV-1 using plasma collected from four distinct cohorts: individuals who received three doses of BBIBP-CorV/CoronaVac vaccines, those who experienced BA.5 breakthrough infections, those with XBB breakthrough infections, and those with BA.5-XBB consecutive infections following three-dose vaccination. FINDINGS Following Omicron breakthrough infections, the levels of nAbs against WT and pre-Omicron VOCs were higher due to immune imprinting established by WT-based vaccination, in comparison to nAbs against Omicron variants. Interestingly, the XBB breakthrough infections elicited a broader neutralization spectrum against SARS-CoV-2 variants compared to the BA.5 breakthrough infections. This observation suggests that the XBB variant demonstrates superior immunogenicity relative to BA.5. Notably, hybrid immunization of BA.5 breakthrough infections after WT vaccination led to additional immune imprinting, resulting in a broadened neutralization profile against both WT and BA.5 variants in BA.5-XBB consecutive infections. However, the duration of nAbs was shorter in these reinfections compared to the breakthrough infections. Additionally, the expanded immune imprinting from previous WT vaccination and BA.5 breakthrough infections account for the enhanced plasma neutralization immunodominance observed in the antigenic cartography for BA.5-XBB consecutive infections. INTERPRETATION Overall, we demonstrated a persistent and expanded effect of immune imprinting from prior SARS-CoV-2 exposures. Thus, future vaccines should specifically address the latest variants, and booster shots should be given at a longer interval after the previous infection or vaccination.
Collapse
Affiliation(s)
- Tingting Jia
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Fuxiang Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Yihao Chen
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, PR China
| | - Guancheng Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Qiuyi Xu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Jiamin Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Jiani Wu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Nina Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Liangliang Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Lifang Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Dongli Wang
- Guangming District Center for Disease Control and Prevention, Shenzhen, PR China
| | - Qian Xie
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Chuming Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Sun Yat-sen University, Shenzhen, PR China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, Guangdong, PR China
| | - Yongkun Chen
- Guangdong Provincial Key Laboratory of Infection Immunity and Inflammation, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, PR China.
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, PR China.
| |
Collapse
|
28
|
Akerman A, Fichter C, Milogiannakis V, Esneau C, Silva MR, Ison T, Lopez JA, Naing Z, Caguicla J, Amatayakul-Chantler S, Roth N, Manni S, Hauser T, Barnes T, Boss T, Condylios A, Yeang M, Sato K, Bartlett NN, Darley D, Matthews G, Stark DJ, Promsri S, Rawlinson WD, Murrell B, Kelleher AD, Dwyer D, Sintchenko V, Kok J, Ellis S, Marris K, Knight E, Hoad VC, Irving DO, Gosbell I, Brilot F, Wood J, Aggarwal A, Turville SG. Cross-sectional and longitudinal genotype to phenotype surveillance of SARS-CoV-2 variants over the first four years of the COVID-19 pandemic. EBioMedicine 2024; 110:105415. [PMID: 39549677 PMCID: PMC11599457 DOI: 10.1016/j.ebiom.2024.105415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Continued phenotyping and ongoing molecular epidemiology are important in current and future monitoring of emerging SARS-CoV-2 lineages. Herein we developed pragmatic strategies to track the emergence, spread and phenotype of SARS-CoV-2 variants in Australia in an era of decreasing diagnostic PCR testing and focused cohort-based studies. This was aligned to longitudinal studies that span 4 years of the COVID-19 pandemic. METHODS Throughout 2023, we partnered with diagnostic pathology providers and pathogen genomics teams to identify relevant emerging or circulating variants in the New South Wales (NSW) community. We monitored emerging variants through viral culture, growth algorithms, neutralisation responses and changing entry requirements defined by ACE2 and TMPRSS2 receptor use. To frame this in the context of the pandemic stage, we continued to longitudinally track neutralisation responses at the population level using pooled Intravenous Immunoglobulins (IVIG) derived from in excess of 700,000 donations. FINDINGS In antibodies derived from recent individual donations and thousands of donations pooled in IVIGs, we observed continued neutralisation across prior and emerging variants with EG.5.1, HV.1, XCT and JN.1 ranked as the most evasive SARS-CoV-2 variants. Changes in the type I antibody site at Spike positions 452, 455 and 456 were associated with lowered neutralisation responses in XBB lineages. In longitudinal tracking of population immunity spanning three years, we observed continued maturation of neutralisation breadth to all SARS-CoV-2 variants over time. Whilst neutralisation responses initially displayed high levels of imprinting towards Ancestral and early pre-Omicron lineages, this was slowly countered by increased cross reactive breadth to all variants. We predicted JN.1 to have a marked transmission advantage in late 2023 and this eventuated globally at the start of 2024. We could not attribute this advantage to neutralisation resistance but rather propose that this growth advantage arises from the preferential utilisation of ACE2 pools that cannot engage TMPRSS2 at its Collectrin-Like Domain (CLD). INTERPRETATION The emergence of many SARS-CoV-2 lineages documented at the end of 2023 was found to be initially associated with lowered neutralisation responses. This continued to be countered by the gradual maturation of cross-reactive neutralisation responses over time. The later appearance and dominance of the divergent JN.1 lineage cannot be attributed to a lack of neutralisation responses alone, and our data supports that its dominance is a culmination of both lowered neutralisation and changes in ACE2/TMPRSS2 entry preferences. FUNDING This work was primarily supported by Australian Medical Foundation research grants MRF2005760 (ST, GM & WDR), MRF2001684 (ADK and ST) and Medical Research Future Fund Antiviral Development Call grant (WDR), Medical Research Future Fund COVID-19 grant (MRFF2001684, ADK & SGT) and the New South Wales Health COVID-19 Research Grants Round 2 (SGT).
Collapse
Affiliation(s)
- Anouschka Akerman
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Christina Fichter
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vanessa Milogiannakis
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Camille Esneau
- Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Mariana Ruiz Silva
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Tim Ison
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Joseph A Lopez
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Zin Naing
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | - Joanna Caguicla
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | | | - Nathan Roth
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Sandro Manni
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Thomas Hauser
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Thomas Barnes
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Tino Boss
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Anna Condylios
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | - Malinna Yeang
- Serology and Virology Division (SAViD), NSW HP SEALS, Randwick, Australia
| | - Kenta Sato
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Nathan N Bartlett
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - David Darley
- St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Gail Matthews
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia; St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Damien J Stark
- Molecular Diagnostic Medicine Laboratory, Sydpath, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Susan Promsri
- Histopath, Pinnacle Office Park, Building B, Level 2/4 Drake Ave, Macquarie Park NSW, 2113, Australia
| | | | - Benjamin Murrell
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Anthony D Kelleher
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Dominic Dwyer
- Centre for Infectious Diseases & Microbiology - Public Health and Institute of Clinical Pathology & Medical Research (ICPMR), New South Wales Health Pathology, Westmead, New South Wales, 2145, Australia
| | - Vitali Sintchenko
- Centre for Infectious Diseases & Microbiology - Public Health and Institute of Clinical Pathology & Medical Research (ICPMR), New South Wales Health Pathology, Westmead, New South Wales, 2145, Australia
| | - Jen Kok
- Centre for Infectious Diseases & Microbiology - Public Health and Institute of Clinical Pathology & Medical Research (ICPMR), New South Wales Health Pathology, Westmead, New South Wales, 2145, Australia
| | - Sally Ellis
- New South Wales Ministry of Health, St Leonards, Australia
| | - Kelsi Marris
- New South Wales Ministry of Health, St Leonards, Australia
| | | | - Veronic C Hoad
- Australian Red Cross Lifeblood, Melbourne, Victoria, Australia
| | - David O Irving
- Australian Red Cross Lifeblood, Melbourne, Victoria, Australia
| | - Iain Gosbell
- Australian Red Cross Lifeblood, Melbourne, Victoria, Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - James Wood
- School of Population Health, UNSW Sydney, Kensington, New South Wales, 2052, Australia
| | - Anupriya Aggarwal
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Stuart G Turville
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
29
|
Yisimayi A, Song W, Wang J, Jian F, Yu Y, Chen X, Xu Y, An R, Wang Y, Wang J, Sun H, Wang P, Yu L, Shao F, Jin R, Shen Z, Wang Y, Cao Y. Prolonged Omicron-specific B cell maturation alleviates immune imprinting induced by SARS-CoV-2 inactivated vaccine. Emerg Microbes Infect 2024; 13:2412623. [PMID: 39360822 PMCID: PMC11486138 DOI: 10.1080/22221751.2024.2412623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
SARS-CoV-2 ancestral strain-induced immune imprinting poses great challenges to updating vaccines for new variants. Studies showed that repeated Omicron exposures could override immune imprinting induced by inactivated vaccines but not mRNA vaccines, a disparity yet to be understood. Here, we analyzed the immune imprinting alleviation in inactivated vaccine (CoronaVac) cohorts after a long-term period following breakthrough infections (BTI). We observed in CoronaVac-vaccinated individuals who experienced BA.5/BF.7 BTI, the proportion of Omicron-specific memory B cells (MBCs) substantially increased after an extended period post-Omicron BTI, with their antibodies displaying enhanced somatic hypermutation and neutralizing potency. Consequently, the neutralizing antibody epitope distribution encoded by MBCs post-BA.5/BF.7 BTI after prolonged maturation closely mirrors that in BA.5/BF.7-infected unvaccinated individuals. Together, these results indicate the activation and expansion of Omicron-specific naïve B cells generated by first-time Omicron exposure helped to alleviate CoronaVac-induced immune imprinting, and the absence of this process should have caused the persistent immune imprinting seen in mRNA vaccine recipients.
Collapse
Affiliation(s)
- Ayijiang Yisimayi
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
| | - Weiliang Song
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
| | - Jing Wang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
| | - Fanchong Jian
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
| | - Yuanling Yu
- Changping Laboratory, Beijing, People’s Republic of China
| | - Xiaosu Chen
- Institute for Immunology, College of Life Sciences, Nankai University, Tianjin, People’s Republic of China
| | - Yanli Xu
- Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Ran An
- Changping Laboratory, Beijing, People’s Republic of China
| | - Yao Wang
- Changping Laboratory, Beijing, People’s Republic of China
| | - Jing Wang
- Changping Laboratory, Beijing, People’s Republic of China
| | - Haiyan Sun
- Changping Laboratory, Beijing, People’s Republic of China
| | - Peng Wang
- Changping Laboratory, Beijing, People’s Republic of China
| | - Lingling Yu
- Changping Laboratory, Beijing, People’s Republic of China
| | - Fei Shao
- Changping Laboratory, Beijing, People’s Republic of China
| | - Ronghua Jin
- Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhongyang Shen
- Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, People’s Republic of China
| | - Youchun Wang
- Changping Laboratory, Beijing, People’s Republic of China
- Institute of Medical Biotechnology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, People’s Republic of China
| | - Yunlong Cao
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, People’s Republic of China
- Changping Laboratory, Beijing, People’s Republic of China
- Peking–Tsinghua Center for Life Sciences, Peking University, Beijing, People’s Republic of China
| |
Collapse
|
30
|
Renner TM, Stuible M, Cass B, Perret S, Guimond J, Lord-Dufour S, McCluskie MJ, Durocher Y, Akache B. Reduced cross-protective potential of Omicron compared to ancestral SARS-CoV-2 spike vaccines against potentially zoonotic coronaviruses. NPJ VIRUSES 2024; 2:58. [PMID: 40295830 PMCID: PMC11721134 DOI: 10.1038/s44298-024-00067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/19/2024] [Indexed: 04/30/2025]
Abstract
The COVID-19 pandemic has emphasised the importance of vaccines and preparedness against viral threats crossing species barriers. In response, a worldwide vaccination campaign targeting SARS-CoV-2 was implemented, which provides some cross-protective immunological memory to other coronavirus species with zoonotic potential. Following a vaccination regimen against SARS-CoV-2 spike in a preclinical mouse model, we were able to demonstrate the induction of neutralizing antibodies towards multiple human ACE2 (hACE2)-binding Sarbecovirus spikes. Importantly, compared to vaccines based on the SARS-CoV-2 Reference strain, vaccines based on Omicron spike sequences induced drastically less broadly cross-protective neutralizing antibodies against other hACE2-binding sarbecoviruses. This observation remained true whether the vaccination regimens were based on protein subunit or mRNA / LNP vaccines. Overall, while it may be necessary to update vaccine antigens to combat the evolving SARS-CoV-2 virus for enhanced protection from COVID-19, Reference-based vaccines may be a more valuable tool to protect against novel coronavirus zoonoses.
Collapse
Affiliation(s)
- Tyler M Renner
- National Research Council Canada, Human Health Therapeutics, Ottawa, ON, Canada
| | - Matthew Stuible
- National Research Council Canada, Human Health Therapeutics, Montreal, QC, Canada
| | - Brian Cass
- National Research Council Canada, Human Health Therapeutics, Montreal, QC, Canada
| | - Sylvie Perret
- National Research Council Canada, Human Health Therapeutics, Montreal, QC, Canada
| | - Julie Guimond
- National Research Council Canada, Human Health Therapeutics, Montreal, QC, Canada
| | - Simon Lord-Dufour
- National Research Council Canada, Human Health Therapeutics, Montreal, QC, Canada
| | - Michael J McCluskie
- National Research Council Canada, Human Health Therapeutics, Ottawa, ON, Canada
| | - Yves Durocher
- National Research Council Canada, Human Health Therapeutics, Montreal, QC, Canada.
| | - Bassel Akache
- National Research Council Canada, Human Health Therapeutics, Ottawa, ON, Canada.
| |
Collapse
|
31
|
Belik M, Reinholm A, Kolehmainen P, Heroum J, Maljanen S, Altan E, Österlund P, Laine L, Ritvos O, Pasternack A, Naves RA, Iakubovskaia A, Barkoff AM, He Q, Lempainen J, Tähtinen PA, Ivaska L, Jalkanen P, Julkunen I, Kakkola L. Long-term COVID-19 vaccine- and Omicron infection-induced humoral and cell-mediated immunity. Front Immunol 2024; 15:1494432. [PMID: 39640263 PMCID: PMC11617562 DOI: 10.3389/fimmu.2024.1494432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Mutations occurring in the spike (S) protein of SARS-CoV-2 enables the virus to evade COVID-19 vaccine- and infection-induced immunity. Methods Here we provide a comprehensive analysis of humoral and cell-mediated immunity in 111 healthcare workers who received three or four vaccine doses and were followed up to 12 and 6 months, respectively, after the last vaccine dose. Omicron breakthrough infection occurred in 71% of the vaccinees, enabling evaluation of vaccine- and vaccine/infection-induced hybrid immunity. Results Neutralizing antibodies were the highest against the ancestral D614G and were sequentially reduced against the Omicron variants BA.2, BA.5 and XBB.1.5. S1-specific IgG and neutralizing antibody levels were significantly higher in infected than in uninfected vaccinees, and the fourth vaccine dose in combination with a breakthrough infection resulted in high neutralizing antibody levels against all variants. T cell-mediated immunity, instead, was well retained already after two vaccine doses, and was not significantly strengthened by additional booster vaccine doses or Omicron breakthrough infections. Discussion While humoral immunity is sensitive to mutations in the S protein and thus declined rapidly, the cell-mediated immunity is durable to antigenic variation, which may explain the good efficacy of COVID-19 vaccines against a severe disease.
Collapse
Affiliation(s)
- Milja Belik
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Arttu Reinholm
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Jemna Heroum
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sari Maljanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Eda Altan
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pamela Österlund
- Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Larissa Laine
- Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Olli Ritvos
- Department of Physiology, Medicum, University of Helsinki, Helsinki, Finland
| | - Arja Pasternack
- Department of Physiology, Medicum, University of Helsinki, Helsinki, Finland
| | - Rauno A. Naves
- Department of Physiology, Medicum, University of Helsinki, Helsinki, Finland
| | - Alina Iakubovskaia
- Department of Physiology, Medicum, University of Helsinki, Helsinki, Finland
| | | | - Qiushui He
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFlames Research Flagship Center, University of Turku, Turku, Finland
| | - Johanna Lempainen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Paula A. Tähtinen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Lauri Ivaska
- InFlames Research Flagship Center, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Pinja Jalkanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFlames Research Flagship Center, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Laura Kakkola
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| |
Collapse
|
32
|
Wang W, Bhushan G, Paz S, Stauft CB, Selvaraj P, Goguet E, Bishop-Lilly KA, Subramanian R, Vassell R, Lusvarghi S, Cong Y, Agan B, Richard SA, Epsi NJ, Fries A, Fung CK, Conte MA, Holbrook MR, Wang TT, Burgess TH, Pollett SD, Mitre E, Katzelnick LC, Weiss CD. Human and hamster sera correlate well in identifying antigenic drift among SARS-CoV-2 variants, including JN.1. J Virol 2024; 98:e0094824. [PMID: 39365051 DOI: 10.1128/jvi.00948-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/14/2024] [Indexed: 10/05/2024] Open
Abstract
Antigenic assessments of SARS-CoV-2 variants inform decisions to update COVID-19 vaccines. Primary infection sera are often used for assessments, but such sera are rare due to population immunity from SARS-CoV-2 infections and COVID-19 vaccinations. Here, we show that neutralization titers and breadth of matched human and hamster pre-Omicron variant primary infection sera correlate well and generate similar antigenic maps. The hamster antigenic map shows modest antigenic drift among XBB sub-lineage variants, with JN.1 and BA.4/BA.5 variants within the XBB cluster, but with fivefold to sixfold antigenic differences between these variants and XBB.1.5. Compared to sera following only ancestral or bivalent COVID-19 vaccinations, or with post-vaccination infections, XBB.1.5 booster sera had the broadest neutralization against XBB sub-lineage variants, although a fivefold titer difference was still observed between JN.1 and XBB.1.5 variants. These findings suggest that antibody coverage of antigenically divergent JN.1 could be improved with a matched vaccine antigen.IMPORTANCEUpdates to COVID-19 vaccine antigens depend on assessing how much vaccine antigens differ antigenically from newer SARS-CoV-2 variants. Human sera from single variant infections are ideal for discriminating antigenic differences among variants, but such primary infection sera are now rare due to high population immunity. It remains unclear whether sera from experimentally infected animals could substitute for human sera for antigenic assessments. This report shows that neutralization titers of variant-matched human and hamster primary infection sera correlate well and recognize variants similarly, indicating that hamster sera can be a proxy for human sera for antigenic assessments. We further show that human sera following an XBB.1.5 booster vaccine broadly neutralized XBB sub-lineage variants but titers were fivefold lower against the more recent JN.1 variant. These findings support updating the current COVID-19 vaccine variant composition and developing a framework for assessing antigenic differences in future variants using hamster primary infection sera.
Collapse
Affiliation(s)
- Wei Wang
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gitanjali Bhushan
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie Paz
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Charles B Stauft
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Prabhuanand Selvaraj
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Emilie Goguet
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Kimberly A Bishop-Lilly
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, Maryland, USA
| | - Rahul Subramanian
- Office of Data Science and Emerging Technologies, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Russell Vassell
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sabrina Lusvarghi
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yu Cong
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Brian Agan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Stephanie A Richard
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Nusrat J Epsi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Anthony Fries
- US Air Force School of Aerospace Medicine, Dayton, Ohio, USA
| | - Christian K Fung
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Matthew A Conte
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Michael R Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Tony T Wang
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Timothy H Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Simon D Pollett
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Carol D Weiss
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
33
|
Seow J, Jefferson GCE, Keegan MD, Yau Y, Snell LB, Doores KJ. Profiling serum immunodominance following SARS-CoV-2 primary and breakthrough infection reveals distinct variant-specific epitope usage and immune imprinting. PLoS Pathog 2024; 20:e1012724. [PMID: 39556615 DOI: 10.1371/journal.ppat.1012724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/03/2024] [Indexed: 11/20/2024] Open
Abstract
Over the course of the COVID-19 pandemic, variants have emerged with increased mutations and immune evasive capabilities. This has led to breakthrough infections (BTI) in vaccinated individuals, with a large proportion of the neutralizing antibody response targeting the receptor binding domain (RBD) of the SARS-CoV-2 Spike glycoprotein. Immune imprinting, where prior exposure of the immune system to an antigen can influence the response to subsequent exposures, and its role in a population with heterogenous exposure histories has important implications in future vaccine design. Here, we develop an accessible approach to map epitope immunodominance of the neutralizing antibody response in sera. By using a panel of mutant Spike proteins in a pseudotyped virus neutralization assay, we observed distinct epitope usage in convalescent donors infected during wave 1, or infected with the Delta, or BA.1 variants, highlighting the antigenic diversity of the variant Spikes. Analysis of longitudinal serum samples taken spanning 3 doses of COVID-19 vaccine and subsequent breakthrough infection, showed the influence of immune imprinting from the ancestral-based vaccine, where reactivation of existing B cells elicited by the vaccine resulted in the enrichment of the pre-existing epitope immunodominance. However, subtle shifts in epitope usage in sera were observed following BTI by Omicron sub-lineage variants. Antigenic distance of Spike, time after last exposure, and number of vaccine boosters may play a role in the persistence of imprinting from the vaccine. This study provides insight into RBD neutralizing epitope usage in individuals with varying exposure histories and has implications for design of future SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Jeffrey Seow
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - George C E Jefferson
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Michael D Keegan
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Yeuk Yau
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Luke B Snell
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
- Centre for Clinical Infection and Diagnostics Research, Department of Infectious Diseases, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Katie J Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
34
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
35
|
Cobey S. Vaccination against rapidly evolving pathogens and the entanglements of memory. Nat Immunol 2024; 25:2015-2023. [PMID: 39384979 DOI: 10.1038/s41590-024-01970-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/26/2024] [Indexed: 10/11/2024]
Abstract
Immune memory determines infection risk and responses to future infections and vaccinations over potentially decades of life. Despite its centrality, the dynamics of memory to antigenically variable pathogens remains poorly understood. This Review examines how past exposures shape B cell responses to vaccinations with influenza and SARS-CoV-2. An overriding feature of vaccinations with these pathogens is the recall of primary responses, often termed 'imprinting' or 'original antigenic sin'. These recalled responses can inhibit the generation of new responses unless some incompletely defined conditions are met. Depending on the context, immune memory can increase or decrease the total neutralizing antibody response to variant antigens, with apparent consequences for protection. These effects are easier to measure experimentally than epidemiologically, but there is evidence that both early and recent exposures influence vaccine effectiveness. A few immunological interactions between adaptive immune responses and antigens might explain the seemingly discrepant effects of memory. Overall, the complex observations point to a need for more quantitative approaches to integrate high-dimensional immune data from populations with diverse exposure histories. Such approaches could help identify optimal vaccination strategies against antigenically diverse pathogens.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
36
|
Fryer HA, Geers D, Gommers L, Zaeck LM, Tan NH, Jones-Freeman B, Goorhuis A, Postma DF, Visser LG, Hogarth PM, Koopmans MPG, GeurtsvanKessel CH, O'Hehir RE, van der Kuy PHM, de Vries RD, van Zelm MC. Fourth dose bivalent COVID-19 vaccines outperform monovalent boosters in eliciting cross-reactive memory B cells to Omicron subvariants. J Infect 2024; 89:106246. [PMID: 39127451 DOI: 10.1016/j.jinf.2024.106246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Bivalent COVID-19 vaccines comprising ancestral Wuhan-Hu-1 (WH1) and the Omicron BA.1 or BA.5 subvariant elicit enhanced serum antibody responses to emerging Omicron subvariants. Here, we characterized the RBD-specific memory B cell (Bmem) response following a fourth dose with a BA.1 or BA.5 bivalent vaccine, in direct comparison with a WH1 monovalent fourth dose. Healthcare workers previously immunized with mRNA or adenoviral vector monovalent vaccines were sampled before and one month after a fourth dose with a monovalent or a BA.1 or BA.5 bivalent vaccine. Serum neutralizing antibodies (NAb) were quantified, as well as RBD-specific Bmem with an in-depth spectral flow cytometry panel including recombinant RBD proteins of the WH1, BA.1, BA.5, BQ.1.1, and XBB.1.5 variants. Both bivalent vaccines elicited higher NAb titers against Omicron subvariants compared to the monovalent vaccine. Following either vaccine type, recipients had slightly increased WH1 RBD-specific Bmem numbers. Both bivalent vaccines significantly increased WH1 RBD-specific Bmem binding of all Omicron subvariants tested by flow cytometry, while recognition of Omicron subvariants was not enhanced following monovalent vaccination. IgG1+ Bmem dominated the response, with substantial IgG4+ Bmem only detected in recipients of an mRNA vaccine for their primary dose. Thus, Omicron-based bivalent vaccines can significantly boost NAb and Bmem specific for ancestral WH1 and Omicron variants and improve recognition of descendent subvariants by pre-existing, WH1-specific Bmem beyond that of a monovalent vaccine. This provides new insights into the capacity of variant-based mRNA booster vaccines to improve immune memory against emerging SARS-CoV-2 variants and potentially protect against severe disease. ONE-SENTENCE SUMMARY: Omicron BA.1 and BA.5 bivalent COVID-19 boosters, used as a fourth dose, increase RBD-specific Bmem cross-recognition of Omicron subvariants, both those encoded by the vaccines and antigenically distinct subvariants, further than a monovalent booster.
Collapse
Affiliation(s)
- Holly A Fryer
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Daryl Geers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Lennert Gommers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Luca M Zaeck
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ngoc H Tan
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Bernadette Jones-Freeman
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Abraham Goorhuis
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam, the Netherlands; Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Amsterdam, the Netherlands
| | - Douwe F Postma
- Department of Internal Medicine and Infectious Diseases, University Medical Center Groningen, Groningen, the Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - P Mark Hogarth
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Marion P G Koopmans
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | | - Robyn E O'Hehir
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia
| | - P Hugo M van der Kuy
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Rory D de Vries
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia; Dept. Immunology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
37
|
Nilles EJ, Roberts K, de St Aubin M, Mayfield H, Restrepo AC, Garnier S, Abdalla G, Etienne MC, Duke W, Dumas D, Jarolim P, Oasan T, Peña F, Lopez B, Cruz LDL, Sanchez IM, Murray K, Baldwin M, Skewes-Ramm R, Paulino CT, Lau CL, Kucharski A. Convergence of SARS-CoV-2 spike antibody levels to a population immune setpoint. EBioMedicine 2024; 108:105319. [PMID: 39232463 PMCID: PMC11404201 DOI: 10.1016/j.ebiom.2024.105319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/06/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Individual immune responses to SARS-CoV-2 are well-studied, while the combined effect of these responses on population-level immune dynamics remains poorly understood. Given the key role of population immunity on pathogen transmission, delineation of the factors that drive population immune evolution has critical public health implications. METHODS We enrolled individuals 5 years and older selected using a multistage cluster survey approach in the Northwest and Southeast of the Dominican Republic. Paired blood samples were collected mid-pandemic (Aug 2021) and late pandemic (Nov 2022). We measured serum pan-immunoglobulin antibodies against the SARS-CoV-2 spike protein. Generalized Additive Models (GAMs) and random forest models were used to analyze the relationship between changes in antibody levels and various predictor variables. Principal component analysis and partial dependence plots further explored the relationships between predictors and antibody changes. FINDINGS We found a transformation in the distribution of antibody levels from an irregular to a normalized single peak Gaussian distribution that was driven by titre-dependent boosting. This led to the convergence of antibody levels around a common immune setpoint, irrespective of baseline titres and vaccination profile. INTERPRETATION Our results suggest that titre-dependent kinetics driven by widespread transmission direct the evolution of population immunity in a consistent manner. These findings have implications for targeted vaccination strategies and improved modeling of future transmission, providing a preliminary blueprint for understanding population immune dynamics that could guide public health and vaccine policy for SARS-CoV-2 and potentially other pathogens. FUNDING The study was primarily funded by the Centers for Disease Control and Prevention grant U01GH002238 (EN). Salary support was provided by Wellcome Trust grant 206250/Z/17/Z (AK) and the Australian National Health and Medical Research Council Investigator grant APP1158469 (CLL).
Collapse
Affiliation(s)
- Eric J Nilles
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA.
| | - Kathryn Roberts
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | - Michael de St Aubin
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | | | | | - Salome Garnier
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | | | | | - William Duke
- Pedro Henríquez Ureña National University, Santo Domingo, Dominican Republic
| | - Devan Dumas
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | - Petr Jarolim
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Farah Peña
- Ministry of Health and Social Assistance, Santo Domingo, Dominican Republic
| | - Beatriz Lopez
- Centers for Disease Control and Prevention, Central America Regional Office, Guatemala City, Guatemala
| | - Lucia de la Cruz
- Ministry of Health and Social Assistance, Santo Domingo, Dominican Republic
| | | | - Kristy Murray
- Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Margaret Baldwin
- Brigham and Womens Hospital, Boston, MA, USA; Harvard Humanitarian Initiative, Cambridge, MA, USA
| | - Ronald Skewes-Ramm
- Ministry of Health and Social Assistance, Santo Domingo, Dominican Republic
| | | | | | - Adam Kucharski
- London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
38
|
Paciello I, Pierleoni G, Pantano E, Antonelli G, Pileri P, Maccari G, Cardamone D, Realini G, Perrone F, Neto MM, Pozzessere S, Fabbiani M, Panza F, Rancan I, Tumbarello M, Montagnani F, Medini D, Maes P, Temperton N, Simon-Loriere E, Schwartz O, Rappuoli R, Andreano E. Antigenic sin and multiple breakthrough infections drive converging evolution of COVID-19 neutralizing responses. Cell Rep 2024; 43:114645. [PMID: 39207904 PMCID: PMC11422482 DOI: 10.1016/j.celrep.2024.114645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Understanding the evolution of the B cell response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants is fundamental to design the next generation of vaccines and therapeutics. We longitudinally analyze at the single-cell level almost 900 neutralizing human monoclonal antibodies (nAbs) isolated from vaccinated people and from individuals with hybrid and super hybrid immunity (SH), developed after three mRNA vaccine doses and two breakthrough infections. The most potent neutralization and Fc functions against highly mutated variants belong to the SH cohort. Repertoire analysis shows that the original Wuhan antigenic sin drives the convergent expansion of the same B cell germlines in vaccinated and SH cohorts. Only Omicron breakthrough infections expand previously unseen germ lines and generate broadly nAbs by restoring IGHV3-53/3-66 germ lines. Our analyses find that B cells initially expanded by the original antigenic sin continue to play a fundamental role in the evolution of the immune response toward an evolving virus.
Collapse
Affiliation(s)
- Ida Paciello
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Giulio Pierleoni
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy; Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Siena, Italy
| | - Elisa Pantano
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy; Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Siena, Italy
| | - Giada Antonelli
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Piero Pileri
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Giuseppe Maccari
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Dario Cardamone
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Giulia Realini
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Federica Perrone
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy; Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Siena, Italy
| | - Martin Mayora Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Simone Pozzessere
- Department of Cellular Therapies, Hematology, and Laboratory Medicine, University Hospital of Siena, Siena, Italy
| | - Massimiliano Fabbiani
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, Siena University Hospital, Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Panza
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, Siena University Hospital, Siena, Italy
| | - Ilaria Rancan
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, Siena University Hospital, Siena, Italy
| | - Mario Tumbarello
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, Siena University Hospital, Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesca Montagnani
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, Siena University Hospital, Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Duccio Medini
- Data Science for Health (DaScH) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | - Piet Maes
- KU Leuven, Rega Institute, Department of Microbiology, Immunology, and Transplantation, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Etienne Simon-Loriere
- G5 Evolutionary Genomics of RNA Viruses, Institut Pasteur, Université Paris Cité, Paris, France; National Reference Center for Respiratory Viruses, Institut Pasteur, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, Paris, France; Vaccine Research Institute, Creteil, France
| | - Rino Rappuoli
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Siena, Italy; Fondazione Biotecnopolo di Siena, Siena, Italy
| | - Emanuele Andreano
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy.
| |
Collapse
|
39
|
Ying B, Liang CY, Desai P, Scheaffer SM, Elbashir SM, Edwards DK, Thackray LB, Diamond MS. Ipsilateral or contralateral boosting of mice with mRNA vaccines confers equivalent immunity and protection against a SARS-CoV-2 Omicron strain. J Virol 2024; 98:e0057424. [PMID: 39194250 PMCID: PMC11406931 DOI: 10.1128/jvi.00574-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Boosting with mRNA vaccines encoding variant-matched spike proteins has been implemented to mitigate their reduced efficacy against emerging SARS-CoV-2 variants. Nonetheless, in humans, it remains unclear whether boosting in the ipsilateral or contralateral arm with respect to the priming doses impacts immunity and protection. Here, we boosted K18-hACE2 mice with either monovalent mRNA-1273 (Wuhan-1 spike) or bivalent mRNA-1273.214 (Wuhan-1 + BA.1 spike) vaccine in the ipsilateral or contralateral leg after a two-dose priming series with mRNA-1273. Boosting in the ipsilateral or contralateral leg elicited equivalent levels of serum IgG and neutralizing antibody responses against Wuhan-1 and BA.1. While contralateral boosting with mRNA vaccines resulted in the expansion of spike-specific B and T cells beyond the ipsilateral draining lymph node (DLN) to the contralateral DLN, administration of a third mRNA vaccine dose at either site resulted in similar levels of antigen-specific germinal center B cells, plasmablasts/plasma cells, T follicular helper cells, and CD8+ T cells in the DLNs and the spleen. Furthermore, ipsilateral and contralateral boosting with mRNA-1273 or mRNA-1273.214 vaccines conferred similar homologous or heterologous immune protection against SARS-CoV-2 BA.1 virus challenge with equivalent reductions in viral RNA and infectious virus in the nasal turbinates and lungs. Collectively, our data show limited differences in B and T cell immune responses after ipsilateral and contralateral site boosting by mRNA vaccines that do not substantively impact protection against an Omicron strain.IMPORTANCESequential boosting with mRNA vaccines has been an effective strategy to overcome waning immunity and neutralization escape by emerging SARS-CoV-2 variants. However, it remains unclear how the site of boosting relative to the primary vaccination series shapes optimal immune responses or breadth of protection against variants. In K18-hACE2 transgenic mice, we observed that intramuscular boosting with historical monovalent or variant-matched bivalent vaccines in the ipsilateral or contralateral limb elicited comparable levels of serum spike-specific antibody and antigen-specific B and T cell responses. Moreover, boosting on either side conferred equivalent protection against a SARS-CoV-2 Omicron challenge strain. Our data in mice suggest that the site of intramuscular boosting with an mRNA vaccine does not substantially impact immunity or protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Baoling Ying
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chieh-Yu Liang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
40
|
Eshraghi R, Bahrami A, Karimi Houyeh M, Nasr Azadani M. JN.1 and the ongoing battle: unpacking the characteristics of a new dominant COVID-19 variant. Pathog Glob Health 2024; 118:453-458. [PMID: 38884317 PMCID: PMC11441051 DOI: 10.1080/20477724.2024.2369378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024] Open
Abstract
In the fourth year of the COVID-19 occurrence, a new COVID-19 variant, JN.1, has emerged and spread globally and become the dominant strain in several regions. It has some specific mutations in its spike proteins, empowering it with higher transmissibility. Regarding the significance of the issue, understanding the clinical and immunological traits of JN.1 is critical for enhancing health strategies and vaccination efforts globally, with the ultimate goal of bolstering our collective response to the pandemic. In this study, we take a look at the latest findings of JN.1 characteristics and mutations as well as its consequences on bypassing immune system. We demonstrate the importance of continual surveillance and strategic adaptation within healthcare frameworks along with the significance of wastewater sampling for the rapid identification of emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
| | - Ashkan Bahrami
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Maryam Nasr Azadani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
41
|
Muik A, Quandt J, Lui BG, Bacher M, Lutz S, Grünenthal M, Toker A, Grosser J, Ozhelvaci O, Blokhina O, Shpyro S, Vogler I, Salisch N, Türeci Ö, Sahin U. Immunity against conserved epitopes dominates after two consecutive exposures to SARS-CoV-2 Omicron BA.1. Cell Rep 2024; 43:114567. [PMID: 39097927 DOI: 10.1016/j.celrep.2024.114567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/07/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure histories become increasingly complex through original and variant-adapted vaccines and infections with viral variants. Upon exposure to the highly altered Omicron spike glycoprotein, pre-immunized individuals predominantly mount recall responses of Wuhan-Hu-1 (wild-type)-imprinted memory B (BMEM) cells mostly targeting conserved non-neutralizing epitopes, leading to diminished Omicron neutralization. We investigated the impact of imprinting in individuals double/triple vaccinated with a wild-type-strain-based mRNA vaccine who, thereafter, had two consecutive exposures to Omicron BA.1 spike (breakthrough infection followed by BA.1-adapted vaccine). We found that depletion of conserved epitope-recognizing antibodies using a wild-type spike bait results in strongly diminished BA.1 neutralization. Furthermore, spike-specific BMEM cells recognizing conserved epitopes are much more prevalent than BA.1-specific BMEM cells. Our observations suggest that imprinted BMEM cell recall responses limit the induction of strain-specific responses even after two consecutive BA.1 spike exposures. Vaccine adaptation strategies need to consider that prior SARS-CoV-2 infections and vaccinations may cause persistent immune imprinting.
Collapse
Affiliation(s)
| | | | | | - Maren Bacher
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany
| | | | | | - Aras Toker
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany
| | | | | | | | | | | | | | - Özlem Türeci
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany; HI-TRON - Helmholtz Institute for Translational Oncology Mainz by DKFZ, Obere Zahlbacherstr. 63, 55131 Mainz, Germany
| | - Ugur Sahin
- BioNTech, An der Goldgrube 12, 55131 Mainz, Germany; TRON gGmbH - Translational Oncology at the University Medical Center of the Johannes Gutenberg University, Freiligrathstraße 12, 55131 Mainz, Germany.
| |
Collapse
|
42
|
Kotaki R, Moriyama S, Oishi S, Onodera T, Adachi Y, Sasaki E, Ishino K, Morikawa M, Takei H, Takahashi H, Takano T, Nishiyama A, Yumoto K, Terahara K, Isogawa M, Matsumura T, Shinkai M, Takahashi Y. Repeated Omicron exposures redirect SARS-CoV-2-specific memory B cell evolution toward the latest variants. Sci Transl Med 2024; 16:eadp9927. [PMID: 39167666 DOI: 10.1126/scitranslmed.adp9927] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Immunological imprinting by ancestral SARS-CoV-2 strains is thought to impede the robust induction of Omicron-specific humoral responses by Omicron-based booster vaccines. Here, we analyzed the specificity and neutralization activity of memory B (Bmem) cells after repeated BA.5 exposure in individuals previously imprinted by ancestral strain-based mRNA vaccines. After a second BA.5 exposure, Bmem cells with BA.5 spike protein-skewed reactivity were promptly elicited, correlating with preexisting antibody titers. Clonal lineage analysis identified BA.5-skewed Bmem cells that had redirected their specificity from the ancestral strain to BA.5 through somatic hypermutations. Moreover, Bmem cells with redirected BA.5 specificity exhibited accelerated development compared with de novo Bmem cells derived from naïve repertoires. This redirected BA.5 specificity demonstrated greater resilience to viral point mutation and adaptation to recent Omicron variants HK.3 and JN.1, months after the second BA.5 exposure, suggesting that existing Bmem cells elicited by older vaccines can redirect their specificity toward newly evolving variants.
Collapse
Affiliation(s)
- Ryutaro Kotaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Shintaro Oishi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Eita Sasaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kota Ishino
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | | | | | - Tomohiro Takano
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Ayae Nishiyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kohei Yumoto
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kazutaka Terahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Masanori Isogawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | | | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido 001-0021, Japan
| |
Collapse
|
43
|
Kim W. Germinal Center Response to mRNA Vaccination and Impact of Immunological Imprinting on Subsequent Vaccination. Immune Netw 2024; 24:e28. [PMID: 39246619 PMCID: PMC11377948 DOI: 10.4110/in.2024.24.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/29/2024] [Indexed: 09/10/2024] Open
Abstract
Vaccines are the most effective intervention currently available, offering protective immunity against targeted pathogens. The emergence of the coronavirus disease 2019 pandemic has prompted rapid development and deployment of lipid nanoparticle encapsulated, mRNA-based vaccines. While these vaccines have demonstrated remarkable immunogenicity, concerns persist regarding their ability to confer durable protective immunity to continuously evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. This review focuses on human B cell responses induced by SARS-CoV-2 mRNA vaccination, with particular emphasis on the crucial role of germinal center reactions in shaping enduring protective immunity. Additionally, we explored observations of immunological imprinting and dynamics of recalled pre-existing immunity following variants of concern-based booster vaccination. Insights from this review contribute to comprehensive understanding B cell responses to mRNA vaccination in humans, thereby refining vaccination strategies for optimal and sustained protection against evolving coronavirus variants.
Collapse
Affiliation(s)
- Wooseob Kim
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
44
|
Fantin RF, Clark JJ, Cohn H, Jaiswal D, Bozarth B, Civljak A, Rao V, Lobo I, Nardulli JR, Srivastava K, Yong J, Andreata-Santos R, Bushfield K, Lee ES, Singh G, PVI Study Group, Kleinstein SH, Krammer F, Simon V, Bajic G, Coelho CH. Dissecting human monoclonal antibody responses from mRNA- and protein-based XBB.1.5 COVID-19 monovalent vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.15.602781. [PMID: 39071292 PMCID: PMC11275766 DOI: 10.1101/2024.07.15.602781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The emergence of highly contagious and immune-evasive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has required reformulation of coronavirus disease 2019 (COVID-19) vaccines to target those new variants specifically. While previous infections and booster vaccinations can enhance variant neutralization, it is unclear whether the monovalent version, administered using either mRNA or protein-based vaccine platforms, can elicit de novo B-cell responses specific for Omicron XBB.1.5 variants. Here, we dissected the genetic antibody repertoire of 603 individual plasmablasts derived from five individuals who received a monovalent XBB.1.5 vaccination either with mRNA (Moderna or Pfizer/BioNtech) or adjuvanted protein (Novavax). From these sequences, we expressed 100 human monoclonal antibodies and determined binding, affinity and protective potential against several SARS-CoV-2 variants, including JN.1. We then select two vaccine-induced XBB.1.5 mAbs, M2 and M39. M2 mAb was a de novo, antibody, i.e., specific for XBB.1.5 but not ancestral SARS-CoV-2. M39 bound and neutralized both XBB.1.5 and JN.1 strains. Our high-resolution cryo-electron microscopy (EM) structures of M2 and M39 in complex with the XBB.1.5 spike glycoprotein defined the epitopes engaged and revealed the molecular determinants for the mAbs' specificity. These data show, at the molecular level, that monovalent, variant-specific vaccines can elicit functional antibodies, and shed light on potential functional and genetic differences of mAbs induced by vaccinations with different vaccine platforms.\.
Collapse
Affiliation(s)
- Raianna F. Fantin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordan J. Clark
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hallie Cohn
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepika Jaiswal
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bailey Bozarth
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alesandro Civljak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vishal Rao
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Igor Lobo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica R. Nardulli
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeremy Yong
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Andreata-Santos
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Retrovirology Laboratory, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Kaitlyn Bushfield
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edward S. Lee
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - PVI Study Group
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven H. Kleinstein
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
- Program in Computational Biology and Biomedical Informatics, Yale University, New Haven, CT 06520, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Goran Bajic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Camila H. Coelho
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
45
|
Schiepers A, Van't Wout MFL, Hobbs A, Mesin L, Victora GD. Opposing effects of pre-existing antibody and memory T cell help on the dynamics of recall germinal centers. Immunity 2024; 57:1618-1628.e4. [PMID: 38838672 PMCID: PMC11236515 DOI: 10.1016/j.immuni.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/20/2024] [Accepted: 05/10/2024] [Indexed: 06/07/2024]
Abstract
Re-exposure to an antigen generates abundant antibody responses and drives the formation of secondary germinal centers (GCs). Recall GCs in mice consist almost entirely of naïve B cells, whereas recall antibodies derive overwhelmingly from memory B cells. Here, we examine this division between cellular and serum compartments. After repeated immunization with the same antigen, tetramer analyses of recall GCs revealed a marked decrease in the ability of B cells in these structures to bind the antigen. Boosting with viral variant proteins restored antigen binding in recall GCs, as did genetic ablation of primary-derived antibody-secreting cells through conditional deletion of Prdm1, demonstrating suppression of GC recall responses by pre-existing antibodies. In hapten-carrier experiments in which B and T cell specificities were uncoupled, memory T cell help allowed B cells with undetectable antigen binding to access GCs. Thus, antibody-mediated feedback steers recall GC B cells away from previously targeted epitopes and enables specific targeting of variant epitopes, with implications for vaccination protocols.
Collapse
Affiliation(s)
- Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | | | - Alvaro Hobbs
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
46
|
Zhang Y, Chamblee M, Xu J, Qu P, Shamseldin MM, Yoo SJ, Misny J, Thongpan I, Kc M, Hall JM, Gupta YA, Evans JP, Lu M, Ye C, Hsu CC, Liang X, Martinez-Sobrido L, Yount JS, Boyaka PN, Liu SL, Dubey P, Peeples ME, Li J. Three SARS-CoV-2 spike protein variants delivered intranasally by measles and mumps vaccines are broadly protective. Nat Commun 2024; 15:5589. [PMID: 38961063 PMCID: PMC11222507 DOI: 10.1038/s41467-024-49443-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
As the new SARS-CoV-2 Omicron variants and subvariants emerge, there is an urgency to develop intranasal, broadly protective vaccines. Here, we developed highly efficacious, intranasal trivalent SARS-CoV-2 vaccine candidates (TVC) based on three components of the MMR vaccine: measles virus (MeV), mumps virus (MuV) Jeryl Lynn (JL1) strain, and MuV JL2 strain. Specifically, MeV, MuV-JL1, and MuV-JL2 vaccine strains, each expressing prefusion spike (preS-6P) from a different variant of concern (VoC), were combined to generate TVCs. Intranasal immunization of IFNAR1-/- mice and female hamsters with TVCs generated high levels of S-specific serum IgG antibodies, broad neutralizing antibodies, and mucosal IgA antibodies as well as tissue-resident memory T cells in the lungs. The immunized female hamsters were protected from challenge with SARS-CoV-2 original WA1, B.1.617.2, and B.1.1.529 strains. The preexisting MeV and MuV immunity does not significantly interfere with the efficacy of TVC. Thus, the trivalent platform is a promising next-generation SARS-CoV-2 vaccine candidate.
Collapse
Affiliation(s)
- Yuexiu Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Michelle Chamblee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Jiayu Xu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Panke Qu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Mohamed M Shamseldin
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Department of Microbiology, The Ohio State University, Columbus, OH, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan, Egypt
| | - Sung J Yoo
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Jack Misny
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ilada Thongpan
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Mahesh Kc
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Jesse M Hall
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yash A Gupta
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - John P Evans
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Cheng Chih Hsu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | | | - Jacob S Yount
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Shan-Lu Liu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, USA
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA.
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
47
|
Tarke A, Ramezani-Rad P, Alves Pereira Neto T, Lee Y, Silva-Moraes V, Goodwin B, Bloom N, Siddiqui L, Avalos L, Frazier A, Zhang Z, da Silva Antunes R, Dan J, Crotty S, Grifoni A, Sette A. SARS-CoV-2 breakthrough infections enhance T cell response magnitude, breadth, and epitope repertoire. Cell Rep Med 2024; 5:101583. [PMID: 38781962 PMCID: PMC11228552 DOI: 10.1016/j.xcrm.2024.101583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/22/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Little is known about the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 or SARS2) vaccine breakthrough infections (BTIs) on the magnitude and breadth of the T cell repertoire after exposure to different variants. We studied samples from individuals who experienced symptomatic BTIs during Delta or Omicron waves. In the pre-BTI samples, 30% of the donors exhibited substantial immune memory against non-S (spike) SARS2 antigens, consistent with previous undiagnosed asymptomatic SARS2 infections. Following symptomatic BTI, we observed (1) enhanced S-specific CD4 and CD8 T cell responses in donors without previous asymptomatic infection, (2) expansion of CD4 and CD8 T cell responses to non-S targets (M, N, and nsps) independent of SARS2 variant, and (3) generation of novel epitopes recognizing variant-specific mutations. These variant-specific T cell responses accounted for 9%-15% of the total epitope repertoire. Overall, BTIs boost vaccine-induced immune responses by increasing the magnitude and by broadening the repertoire of T cell antigens and epitopes recognized.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Parham Ramezani-Rad
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Yeji Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Vanessa Silva-Moraes
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Benjamin Goodwin
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Nathaniel Bloom
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Leila Siddiqui
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Liliana Avalos
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Zeli Zhang
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Jennifer Dan
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
48
|
Liang CY, Raju S, Liu Z, Li Y, Asthagiri Arunkumar G, Case JB, Scheaffer SM, Zost SJ, Acreman CM, Gagne M, Andrew SF, Carvalho Dos Anjos DC, Foulds KE, McLellan JS, Crowe JE, Douek DC, Whelan SPJ, Elbashir SM, Edwards DK, Diamond MS. Imprinting of serum neutralizing antibodies by Wuhan-1 mRNA vaccines. Nature 2024; 630:950-960. [PMID: 38749479 PMCID: PMC11419699 DOI: 10.1038/s41586-024-07539-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/08/2024] [Indexed: 06/21/2024]
Abstract
Immune imprinting is a phenomenon in which prior antigenic experiences influence responses to subsequent infection or vaccination1,2. The effects of immune imprinting on serum antibody responses after boosting with variant-matched SARS-CoV-2 vaccines remain uncertain. Here we characterized the serum antibody responses after mRNA vaccine boosting of mice and human clinical trial participants. In mice, a single dose of a preclinical version of mRNA-1273 vaccine encoding Wuhan-1 spike protein minimally imprinted serum responses elicited by Omicron boosters, enabling generation of type-specific antibodies. However, imprinting was observed in mice receiving an Omicron booster after two priming doses of mRNA-1273, an effect that was mitigated by a second booster dose of Omicron vaccine. In both SARS-CoV-2-infected and uninfected humans who received two Omicron-matched boosters after two or more doses of the prototype mRNA-1273 vaccine, spike-binding and neutralizing serum antibodies cross-reacted with Omicron variants as well as more distantly related sarbecoviruses. Because serum neutralizing responses against Omicron strains and other sarbecoviruses were abrogated after pre-clearing with Wuhan-1 spike protein, antibodies induced by XBB.1.5 boosting in humans focus on conserved epitopes targeted by the antecedent mRNA-1273 primary series. Thus, the antibody response to Omicron-based boosters in humans is imprinted by immunizations with historical mRNA-1273 vaccines, but this outcome may be beneficial as it drives expansion of cross-neutralizing antibodies that inhibit infection of emerging SARS-CoV-2 variants and distantly related sarbecoviruses.
Collapse
Affiliation(s)
- Chieh-Yu Liang
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Saravanan Raju
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Yuhao Li
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | | | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Seth J Zost
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cory M Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shayne F Andrew
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | | | | | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
49
|
Wang W, Bhushan GL, Paz S, Stauft CB, Selvaraj P, Goguet E, Bishop-Lilly KA, Subramanian R, Vassell R, Lusvarghi S, Cong Y, Agan B, Richard SA, Epsi NJ, Fries A, Fung CK, Conte MA, Holbrook MR, Wang TT, Burgess TH, Mitre E, Pollett SD, Katzelnick LC, Weiss CD. Antigenic cartography using hamster sera identifies SARS-CoV-2 JN.1 evasion seen in human XBB.1.5 booster sera. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588359. [PMID: 38712124 PMCID: PMC11071293 DOI: 10.1101/2024.04.05.588359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Antigenic assessments of SARS-CoV-2 variants inform decisions to update COVID-19 vaccines. Primary infection sera are often used for assessments, but such sera are rare due to population immunity from SARS-CoV-2 infections and COVID-19 vaccinations. Here, we show that neutralization titers and breadth of matched human and hamster pre-Omicron variant primary infection sera correlate well and generate similar antigenic maps. The hamster antigenic map shows modest antigenic drift among XBB sub-lineage variants, with JN.1 and BA.4/BA.5 variants within the XBB cluster, but with five to six-fold antigenic differences between these variants and XBB.1.5. Compared to sera following only ancestral or bivalent COVID-19 vaccinations, or with post-vaccination infections, XBB.1.5 booster sera had the broadest neutralization against XBB sub-lineage variants, although a five-fold titer difference was still observed between JN.1 and XBB.1.5 variants. These findings suggest that antibody coverage of antigenically divergent JN.1 could be improved with a matched vaccine antigen.
Collapse
Affiliation(s)
- Wei Wang
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gitanjali L. Bhushan
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie Paz
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Charles B. Stauft
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Prabhu Selvaraj
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Emilie Goguet
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Bethesda, Maryland, USA
| | - Kimberly A. Bishop-Lilly
- Biological Defense Research Directorate, Naval Medical Research Command, Fort Detrick, Maryland, USA
| | - Rahul Subramanian
- Office of Data Science and Emerging Technologies, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Russell Vassell
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sabrina Lusvarghi
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yu Cong
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, USA
| | - Brian Agan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Stephanie A. Richard
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Nusrat J. Epsi
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Anthony Fries
- US Air Force School of Aerospace Medicine, Dayton, Ohio, USA
| | - Christian K. Fung
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Matthew A. Conte
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Michael R. Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, USA
| | - Tony T. Wang
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Timothy H. Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Edward Mitre
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Simon D. Pollett
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Bethesda, Maryland, USA
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Leah C. Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Carol D. Weiss
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|