1
|
Beaudoin-Bussières G, Tauzin A, Dionne K, El Ferri O, Benlarbi M, Bourassa C, Medjahed H, Bazin R, Côté M, Finzi A. Multiple exposures to SARS-CoV-2 Spike enhance cross-reactive antibody-dependent cellular cytotoxicity against SARS-CoV-1. Virology 2025; 607:110512. [PMID: 40147380 DOI: 10.1016/j.virol.2025.110512] [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: 10/29/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Vaccination or infection by SARS-CoV-2 elicits a protective immune response against severe outcomes. It has been reported that SARS-CoV-2 infection or vaccination elicits cross-reactive antibodies against other betacoronaviruses. While plasma neutralizing capacity was studied in great detail, their Fc-effector functions remain understudied. Here, we analyzed Spike recognition, neutralization and antibody-dependent cellular cytotoxicity (ADCC) against D614G, a recent Omicron subvariant of SARS-CoV-2 (JN.1) and SARS-CoV-1. Plasma from individuals before their first dose of mRNA vaccine, and following their second, third and sixth doses were analyzed. Despite poor neutralization activity observed after the second and third vaccine doses, ADCC was readily detected. By the sixth dose, individuals could neutralize and mediate ADCC against JN.1 and SARS-CoV-1. Since previous reports have shown that Fc-effector functions were associated with survival from acute infection, these results suggest that ADCC could help in combating future SARS-CoV-2 variants as well as closely related coronaviruses.
Collapse
Affiliation(s)
- Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Montréal, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Alexandra Tauzin
- Centre de Recherche du CHUM, Montréal, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Katrina Dionne
- Centre de Recherche du CHUM, Montréal, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | - Omar El Ferri
- Department of Biochemistry, Microbiology and Immunology, and Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Mehdi Benlarbi
- Centre de Recherche du CHUM, Montréal, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada
| | | | | | - Renée Bazin
- Héma-Québec, Affaires Médicales et Innovation, Québec, QC, G1V 5C3, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, and Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montréal, QC, H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H2X 0A9, Canada.
| |
Collapse
|
2
|
Machado A, Kislaya I, Soares P, Magalhães S, Nunes B. Long term bivalent mRNA vaccine effectiveness against COVID-19 hospitalisations and deaths in Portugal: a cohort study based on electronic health records. BMC Infect Dis 2025; 25:590. [PMID: 40269729 PMCID: PMC12020219 DOI: 10.1186/s12879-025-10866-x] [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: 07/26/2024] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND In Autumn 2022, there were recommendations for a COVID-19 booster vaccination with adapted bivalent vaccines to eligible population. Evaluating vaccine effectiveness (VE), in a short period after the vaccination, is key to guide public health decisions on the vaccine performance, allowing implementation of mitigation strategies promptly. However, to assess long-term protection post-vaccination and evaluate the need for additional boosters, it is crucial to conduct studies that span the maximum duration of the vaccination program. This study aims to estimate the VE of bivalent mRNA vaccines against COVID-19-related hospitalisation and death in the Portuguese population aged 65 years or older, from September 2022 to May 2023. METHODS We used a cohort approach to analyse six electronic health registries using deterministic linkage, with a follow-up period of eight months. Severe outcomes included COVID-19-related hospitalisations and death, classified using discharge ICD-10 codes as proxies. The exposure of interest was the bivalent mRNA vaccine. VE was estimated for 14-97, 98-181 and 182-240 days after bivalent vaccination. Confounder-adjusted hazard ratio (aHR) was obtained by fitting a time-dependent Cox regression model with time-dependent vaccination status, adjusted for sociodemographic, history of influenza and pneumococcus vaccination, previous SARS-CoV-2 tests and infection, and comorbidities. VE was estimated by one minus the aHR between vaccinated with bivalent vaccine person-years versus those without bivalent vaccine person-years. RESULTS The cohort included 2,151,531 individuals aged 65 or older (27.8% with 80 or more years). In the ≥ 80 years old, VE was 41.3% (95%CI: 34.5-47.5%) and 50.3% (95%CI: 44.6-55.3%) against COVID-19-related hospitalisation and death, respectively. In the 65-79 years old, VE was 58.5% (95%CI: 51.9-64.2%) against COVID-19-related hospitalisation, and 65.1% (95%CI: 59.0-70.4%) against COVID-19-related death. VE waned for both age groups and outcomes. Among adults aged 65 years or older, we observed long-term moderate VE estimates against severe COVID-19-related outcomes. CONCLUSIONS These results support the need for yearly boosters of COVID-19 vaccination to maximise the protection of the senior population against COVID-19 severe disease. Additional (spring boosters) during a vaccination campaign should be evaluated considering the epidemiological context and results from long-term VE studies.
Collapse
Affiliation(s)
- Ausenda Machado
- Departamento de Epidemiologia, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal.
- Centro de Investigação em Saúde Pública, Universidade NOVA de Lisboa, Lisbon, Portugal.
| | - Irina Kislaya
- Centro de Investigação em Saúde Pública, Universidade NOVA de Lisboa, Lisbon, Portugal
- Department of Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Patricia Soares
- Departamento de Epidemiologia, Instituto Nacional de Saúde Doutor Ricardo Jorge, Lisbon, Portugal
| | - Sarah Magalhães
- Serviços Partilhados do Ministério da Saúde, Lisbon, Portugal
| | - Baltazar Nunes
- Centro de Investigação em Saúde Pública, Universidade NOVA de Lisboa, Lisbon, Portugal
- Epiconcept, Paris, France
| |
Collapse
|
3
|
Sims A, Weir DJ, Cole SJ, Hutchinson E. SARS-CoV-2 cellular coinfection is limited by superinfection exclusion. J Virol 2025; 99:e0207724. [PMID: 40116503 PMCID: PMC11998510 DOI: 10.1128/jvi.02077-24] [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: 11/21/2024] [Accepted: 02/06/2025] [Indexed: 03/23/2025] Open
Abstract
The coinfection of individual cells is a requirement for exchange between two or more virus genomes, which is a major mechanism driving virus evolution. Coinfection is restricted by a mechanism known as superinfection exclusion (SIE), which prohibits the infection of a previously infected cell by a related virus after a period of time. SIE regulates coinfection for many different viruses, but its relevance to the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was unknown. In this study, we investigated this using a pair of SARS-CoV-2 variant viruses encoding distinct fluorescent reporter proteins. We show for the first time that SARS-CoV-2 coinfection of individual cells is limited temporally by SIE. We defined the kinetics of the onset of SIE for SARS-CoV-2 in this system, showing that the potential for coinfection starts to diminish within the first hour of primary infection and then falls exponentially as the time between the two infection events is increased. We then asked how these kinetics would affect the potential for coinfection with viruses during a spreading infection. We used plaque assays to model the localized spread of SARS-CoV-2 observed in infected tissue and showed that the kinetics of SIE restrict coinfection-and therefore sites of possible genetic exchange-to a small interface of infected cells between spreading viral infections. This indicates that SIE, by reducing the likelihood of coinfection of cells, likely reduces the opportunities for genetic exchange between different strains of SARS-CoV-2 and therefore is an underappreciated factor in shaping SARS-CoV-2 evolution. IMPORTANCE Since SARS-CoV-2 first emerged in 2019, it has continued to evolve, occasionally generating variants of concern. One of the ways that SARS-CoV-2 can evolve is through recombination, where genetic information is swapped between different genomes. Recombination requires the coinfection of cells; therefore, factors impacting coinfection are likely to influence SARS-CoV-2 evolution. Coinfection is restricted by SIE, a phenomenon whereby a previously infected cell becomes increasingly resistant to subsequent infection. Here we report that SIE is activated following SARS-CoV-2 infection and reduces the likelihood of coinfection exponentially following primary infection. Furthermore, we show that SIE prevents coinfection of cells at the boundary between two expanding areas of infection, the scenario most likely to lead to recombination between different SARS-CoV-2 lineages. Our work suggests that SIE reduces the likelihood of recombination between SARS-CoV-2 genomes and therefore likely shapes SARS-CoV-2 evolution.
Collapse
Affiliation(s)
- Anna Sims
- MRC-University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | - Daniel J. Weir
- MRC-University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | - Sarah J. Cole
- MRC-University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | - Edward Hutchinson
- MRC-University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| |
Collapse
|
4
|
Chang HH, Lee YH, Huang KC, Chan DC, Lin YC, Sheng WH, Lee LT, Huang LM. COVID-19 vaccination: 2023 Taiwan Association of Gerontology and Geriatrics (TAGG) consensus statements. J Formos Med Assoc 2025; 124:304-310. [PMID: 38991898 DOI: 10.1016/j.jfma.2024.06.027] [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: 05/28/2024] [Revised: 06/16/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024] Open
Abstract
The COVID-19 pandemic remains challenging due to the rapid evolution of the severe acute respiratory syndrome coronavirus 2. This article discusses recent findings on high-risk groups for COVID-19 mortality and morbidity, along with consensus statements from the 2023 Taiwan Association of Gerontology and Geriatrics (TAGG) meeting. It examines evidence on viral mutation mechanisms, emerging variants, and their implications for vaccination strategies. The article underscores advanced age, immunocompromised status, chronic medical conditions, occupational exposure, and socioeconomic disparities as significant risk factors for severe COVID-19 outcomes. TAGG's consensus emphasizes robust vaccination promotion, prioritizing elderly, and immunocompromised groups, individualized multi-dose regimens for immunocompromised patients, and simplified clinical guidelines. Discussions on global and regional recommendations for regular, variant-adapted boosters highlight the non-seasonal nature of COVID-19. Key agreements include escalating domestic preparedness, implementing vigorous risk-based vaccination, and adapting global guidelines to local contexts. Given ongoing viral evolution, proactive adjustment of vaccination policies is essential. Scientific consensus, tailored recommendations, and rapid knowledge dissemination are vital for optimizing COVID-19 protection among vulnerable groups in Taiwan. This article seeks to inform clinical practice and public health policy by summarizing expert-driven vaccination perspectives.
Collapse
Affiliation(s)
- Hao-Hsiang Chang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Yi-Hsuan Lee
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Kuo-Chin Huang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Ding-Cheng Chan
- Department of Geriatrics and Gerontology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Ying-Chin Lin
- Geriatric Medicine Department, Taipei Medical University-Wanfang Hospital, Taipei, 116, Taiwan
| | - Wang-Huei Sheng
- College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Department of Internal Medicine, National Taiwan University Children's Hospital, Taipei, 100, Taiwan
| | - Long-Teng Lee
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan; College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Taipei Jen-Chi Relief Institution, Taipei, 108, Taiwan.
| | - Li-Min Huang
- College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Department of Pediatrics, National Taiwan University Children's Hospital, Taipei, 100, Taiwan.
| |
Collapse
|
5
|
Sun S, He J, Liu L, Zhu Y, Zhang Q, Qiu Y, Han Y, Xue S, Peng X, Long Y, Lu T, Wu W, Xia A, Zhou Y, Yan Y, Gao Y, Lu L, Sun L, Xie M, Wang Q. Anti-S2 antibodies responsible for the SARS-CoV-2 infection-induced serological cross-reactivity against MERS-CoV and MERS-related coronaviruses. Front Immunol 2025; 16:1541269. [PMID: 40226608 PMCID: PMC11985752 DOI: 10.3389/fimmu.2025.1541269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
Sarbecoviruses, such as SARS-CoV-2, utilize angiotensin-converting enzyme 2 (ACE2) as the entry receptor; while merbecoviruses, such as MERS-CoV, use dipeptidyl peptidase 4 (DPP4) for viral entry. Recently, several MERS-related coronaviruses, NeoCoV and PDF-2180, were reported to use ACE2, the same receptor as SARS-CoV-2, to enter cells, raising the possibility of potential recombination between SARS-CoV-2 and MERS-related coronaviruses within the co-infected ACE2-expressing cells. However, facing this potential recombination risk, the serum and antibody cross-reactivity against MERS/MERS-related coronaviruses after SARS-CoV-2 vaccination and/or infection is still elusive. Here, in this study, we showed that the serological cross-reactivity against MERS/MERS-related S proteins could be induced by SARS-CoV-2 infection but not by inactivated SARS-CoV-2 vaccination. Further investigation revealed that this serum cross-reactivity is due to monoclonals recognizing relatively conserved S2 epitopes, such as fusion peptide and stem helix, but not by antibodies against the receptor-binding domain (RBD), N-terminal domain (NTD) or subdomain-1 (SD1). Some of these anti-S2 cross-reactive mAbs showed cross-neutralizing activity, while none of them exhibited antibody-dependent enhancement (ADE) effect of viral entry in vitro. Together, these results dissected the SARS-CoV-2 infection-induced serological cross-reactivity against MERS/MERS-related coronaviruses, and highlighted the significance of conserved S2 region for the design and development of pan-β-coronaviruses vaccines.
Collapse
Affiliation(s)
- Siyuan Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiaying He
- Microbiological Testing Department, Baoshan District Center for Disease Control and Prevention, Shanghai, China
| | - Luotian Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuzhen Zhu
- Department of Gastroenterology, Jingan District Central Hospitals, Fudan University, Shanghai, China
| | - Qingsong Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yinong Qiu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuru Han
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Xue
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaofang Peng
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiming Long
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tianyu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Anqi Xia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yunjiao Zhou
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yan Yan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yidan Gao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lei Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Minxiang Xie
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Zhou M, Ma J, Fang M, Liu X, Zhang C, Wu K, Ye J, Zhang Y, Yuan Q, Chen R, Chen P, Zhu H, Guan Y, Cheng T, Yuan L, Xia N. Increased pathogenicity and transmissibility in hamsters of all age groups reveal an underestimated perniciousness of severe acute respiratory syndrome coronavirus 2 EG.1 variant. iScience 2025; 28:111875. [PMID: 40034851 PMCID: PMC11872406 DOI: 10.1016/j.isci.2025.111875] [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: 06/06/2024] [Revised: 11/16/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
The evolution and mutation of SARS-CoV-2 is elusive. However, the diverse in vivo pathogenicity and transmissibility of different SARS-CoV-2 Omicron/XBB variants are not well understood. We compared virological attributes of two XBB variants, XBB.1.16 and XBB.1.9.2.1 (EG.1) in new-born, juvenile, adult, middle-aged and senescent Syrian hamsters. In particular, EG.1 has a specific Q613H mutation and causes fatal severe pneumonia in hamsters of all ages. In contrast, all hamsters infected with XBB.1.16 survived and showed milder symptoms. The XBB.1.16 infected hamsters lost significantly less body weight and exhibited lower respiratory viral loads, pro-inflammatory cytokines and lung injury than those with EG.1 infection. In addition, EG.1 is more transmissible than XBB.1.16 in close contact co-housing. Both EG.1 and XBB.1.16 are highly resistant to therapeutic antibodies and convalescent serum. Overall, the unpredictable evolution, global transmission and potential threat of emerging SARS-CoV-2 variants necessitate the updating of prophylactic and therapeutic countermeasures in all age groups.
Collapse
Affiliation(s)
- Ming Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Jian Ma
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Xuan Liu
- Clinical Center for Bio-Therapy, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, Fujian, China
| | - Chang Zhang
- Clinical Center for Bio-Therapy, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, Fujian, China
| | - Kun Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Jianghui Ye
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Yali Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Rirong Chen
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, Shantou, Guangdong, China
| | - Peiwen Chen
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, Shantou, Guangdong, China
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, Shantou, Guangdong, China
| | - Yi Guan
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Guangdong-Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU), Shantou University, Shantou, Guangdong, China
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Lunzhi Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Life Sciences, School of Public Health, Xiang an Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
7
|
Dowgier G, Hobbs A, Greenwood D, Shawe-Taylor M, Stevenson-Leggett P, Bazire J, Penn R, Harvey R, Libri V, Kassiotis G, Gamblin S, Lewis NS, Williams B, Swanton C, Gandhi S, Bauer DLV, Carr EJ, Wall EC, Wu MY. Accurate evaluation of live-virus microneutralisation for SARS-CoV-2 variant JN.1 in the assessment of vaccination and therapeutics. Vaccine 2025; 54:126960. [PMID: 40056806 DOI: 10.1016/j.vaccine.2025.126960] [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: 12/04/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/10/2025]
Abstract
Emerging SARS-CoV-2 variants require rapid assessments of pathogenicity and evasion of existing immunity to inform policy. A crucial component of these assessments is accurate estimation of serum neutralising antibody titres using cultured live virus isolates. Here, we report a comparison of culture methods for Omicron sub-variant JN.1 and the subsequent evaluation of neutralising antibody titres (nAbTs) in recipients of BNT162b2-XBB.1.5 monovalent and the ancestral/BA.4/5 containing bivalent vaccines. We compared culture of JN.1 in either Vero V1 cells or Caco-2 cells, finding culture in Vero V1 either resulted in low-titre stocks or induced crucial mutations at the Spike furin cleavage site (FCS). Using sequence-clean culture stocks generated in Caco-2 cells, we assessed serum samples from 71 healthy adults eligible for a COVID-19 vaccination given as a 5th dose booster in the UK: all participants had detectable nAbs against JN.1 prior to vaccination, with baseline/pre-existing nAbTs between both vaccine groups comparable (p = 0.240). However, nAbTs against JN.1 post-vaccination were 2.6-fold higher for recipients of the monovalent XBB.1.5 vaccine than the BA.4/5 bivalent vaccine (p < 0.001). Further, at clinically relevant concentrations the therapeutic monoclonal antibody Sotrovimab marginally maintains neutralisation of JN.1. Regular re-appraisal of methods and policy outcomes as new variants arise is required to ensure robust data are used to underpin future severity assessments and vaccine strain selection decisions.
Collapse
Affiliation(s)
- Giulia Dowgier
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Agnieszka Hobbs
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - David Greenwood
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom
| | - Marianne Shawe-Taylor
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom
| | - Phoebe Stevenson-Leggett
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - James Bazire
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Rebecca Penn
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Ruth Harvey
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Vincenzo Libri
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom
| | - George Kassiotis
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Department of Infectious Disease, St Mary's Hospital, Imperial College London, London, United Kingdom
| | - Steve Gamblin
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Nicola S Lewis
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Bryan Williams
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom; University College London, London, United Kingdom
| | - Charles Swanton
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; University College London, London, United Kingdom
| | - Sonia Gandhi
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; University College London, London, United Kingdom
| | - David L V Bauer
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Genotype-to-Phenotype 2 Consortium (G2P2-UK), United Kingdom
| | - Edward J Carr
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; University College London, London, United Kingdom
| | - Emma C Wall
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom; Research Department of Infection, Division of Infection and Immunity, University College London, United Kingdom
| | - Mary Y Wu
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom.
| |
Collapse
|
8
|
Tao Y, Ge S. A distribution-guided Mapper algorithm. BMC Bioinformatics 2025; 26:73. [PMID: 40045218 PMCID: PMC11881416 DOI: 10.1186/s12859-025-06085-5] [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: 02/08/2024] [Accepted: 02/14/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The Mapper algorithm is an essential tool for exploring the data shape in topological data analysis. With a dataset as an input, the Mapper algorithm outputs a graph representing the topological features of the whole dataset. This graph is often regarded as an approximation of a Reeb graph of a dataset. The classic Mapper algorithm uses fixed interval lengths and overlapping ratios, which might fail to reveal subtle features of a dataset, especially when the underlying structure is complex. RESULTS In this work, we introduce a distribution-guided Mapper algorithm named D-Mapper, which utilizes the property of the probability model and data intrinsic characteristics to generate density-guided covers and provide enhanced topological features. Moreover, we introduce a metric accounting for both the quality of overlap clustering and extended persistent homology to measure the performance of Mapper-type algorithms. Our numerical experiments indicate that the D-Mapper outperforms the classic Mapper algorithm in various scenarios. We also apply the D-Mapper to a SARS-COV-2 coronavirus RNA sequence dataset to explore the topological structure of different virus variants. The results indicate that the D-Mapper algorithm can reveal both the vertical and horizontal evolutionary processes of the viruses. Our code is available at https://github.com/ShufeiGe/D-Mapper . CONCLUSION The D-Mapper algorithm can generate covers from data based on a probability model. This work demonstrates the power of fusing probabilistic models with Mapper algorithms.
Collapse
Affiliation(s)
- Yuyang Tao
- Institute of Mathematical Sciences, ShanghaiTech University, 393 Middle Huaxia Road, 201210, Shanghai, China
| | - Shufei Ge
- Institute of Mathematical Sciences, ShanghaiTech University, 393 Middle Huaxia Road, 201210, Shanghai, China.
| |
Collapse
|
9
|
Tort LFL, de Araújo MF, Arantes I, Martins JSCC, Gomes M, de Carvalho FC, de Almeida WAF, Caetano BC, Appolinario LR, Pereira EC, Carvalho J, Miyajima F, Wallau GL, Naveca FG, Alves P, Espíndola O, Brasil P, Resende PC, Bello G, Siqueira MM. SARS-CoV-2 Omicron XBB infections boost cross-variant neutralizing antibodies, potentially explaining the observed delay of the JN.1 wave in some Brazilian regions. IJID REGIONS 2025; 14:100503. [PMID: 39845926 PMCID: PMC11750507 DOI: 10.1016/j.ijregi.2024.100503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 01/24/2025]
Abstract
Objectives: The SARS-CoV-2 JN.1 lineage emerged in late 2023 and quickly replaced the XBB lineages, becoming the predominant Omicron variant worldwide in 2024. We estimate the epidemiologic impact of this SARS-CoV-2 lineage replacement in Brazil and we further assessed the cross-reactive neutralizing antibody (NAb) responses in a cohort of convalescent Brazilian patients infected during 2023. Methods We analyzed the evolution of SARS-CoV-2 lineages and severe acute respiratory infection (SARI) cases in Brazil between July 2023 and March 2024. We evaluated the cross-reactive NAb responses to the JN.1 variant in a cohort of convalescent Brazilian patients before and after infection with XBB.1* lineages. Results JN.1 replaced XBB with similar temporal dynamics across all country regions, although its epidemiologic impact varied between locations. The southeastern, southern, and central-western regions experienced a brief XBB wave around October 2023, shortly before the introduction of JN.1, without any immediate upsurge of SARI cases during viral lineage replacement. By contrast, the northeastern and northern regions did not experience an XBB wave in the latter half of 2023 and displayed a rapid surge in SARI cases driven by the emergence of the JN.1. We found that recent XBB infections in the Brazilian population significantly boosted cross-reactive NAb levels against JN.1. Conclusion The XBB wave observed in the second half of 2023 in some Brazilian states likely acted as a booster for population immunity, providing short-term protection against JN.1 infections and delaying the rise of SARI cases in certain regions of the country.
Collapse
Affiliation(s)
- Luis Fernando Lopez Tort
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Laboratory of Molecular Virology, Biological Sciences Department, CENUR Litoral Norte, Universidad de la República, Salto, Uruguay
| | - Mia Ferreira de Araújo
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Ighor Arantes
- Laboratory of Arbovirus and Hemorragic Viruses, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Jéssica SCC Martins
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Marcelo Gomes
- Fiocruz, Presidency, Scientific Computing Program, Group of Analytical Methods in Epidemiological Surveillance, Rio de Janeiro, Brazil
- Department of Transmissible Diseases, General Coordination of Surveillance of Covid-19, Influenza and Other Respiratory Viruses, Secretariat of Health and Environmental Surveillance, Ministry of Health, Brasília, Brazil
| | - Felipe Cotrim de Carvalho
- Department of Transmissible Diseases, General Coordination of Surveillance of Covid-19, Influenza and Other Respiratory Viruses, Secretariat of Health and Environmental Surveillance, Ministry of Health, Brasília, Brazil
| | - Walquiria Aparecida Ferreira de Almeida
- Department of Transmissible Diseases, General Coordination of Surveillance of Covid-19, Influenza and Other Respiratory Viruses, Secretariat of Health and Environmental Surveillance, Ministry of Health, Brasília, Brazil
| | - Braulia Costa Caetano
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Luciana R. Appolinario
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Elisa Calvalcante Pereira
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Jéssica Carvalho
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Fábio Miyajima
- Analytical Competence Molecular Epidemiology Laboratory (ACME), Fundação Oswaldo Cruz (FIOCRUZ), Fortaleza, Ceará, Brazil
| | - Gabriel Luz Wallau
- Department of Entomology & Bioinformatics Center, Aggeu Magalhães Institute, FIOCRUZ, S/N Professor Moraes Rego Ave., Pernambuco, Brazil
- Department of Arbovirology, Bernhard Nocht Institute for Tropical Medicine, Who Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research. National Reference Center for Tropical Infectious Diseases, Hamburg, Germany
| | - Felipe Gomes Naveca
- Laboratory of Arbovirus and Hemorragic Viruses, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
- Laboratory of Ecology of Transmissible Diseases of Amazônia, Leônidas e Maria Deane Institute, Amazonas, Brazil
| | - Pedro Alves
- Laboratory of Immunology of Viral diseases, René Rachou Institute, Minas Gerais, Brazil
| | - Otávio Espíndola
- Laboratory of Clinical Research for Acute Febrile Illnesses, Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Patricia Brasil
- Laboratory of Clinical Research for Acute Febrile Illnesses, Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Paola Cristina Resende
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Gonzalo Bello
- Laboratory of Arbovirus and Hemorragic Viruses, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Marilda Mendonça Siqueira
- Laboratory of Respiratory Viruses, Exanthematous and Enteroviruses and Viral Emergencies, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Lytras S, Lamb KD, Ito J, Grove J, Yuan K, Sato K, Hughes J, Robertson DL. Pathogen genomic surveillance and the AI revolution. J Virol 2025; 99:e0160124. [PMID: 39878472 PMCID: PMC11852828 DOI: 10.1128/jvi.01601-24] [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] [Indexed: 01/31/2025] Open
Abstract
The unprecedented sequencing efforts during the COVID-19 pandemic paved the way for genomic surveillance to become a powerful tool for monitoring the evolution of circulating viruses. Herein, we discuss how a state-of-the-art artificial intelligence approach called protein language models (pLMs) can be used for effectively analyzing pathogen genomic data. We highlight examples of pLMs applied to predicting viral properties and evolution and lay out a framework for integrating pLMs into genomic surveillance pipelines.
Collapse
Affiliation(s)
- Spyros Lytras
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Kieran D. Lamb
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- School of Computing Science, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Jumpei Ito
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Joe Grove
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Ke Yuan
- School of Computing Science, University of Glasgow, Glasgow, Scotland, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
- Cancer Research UK Scotland Institute, Glasgow, Scotland, United Kingdom
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - David L. Robertson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| |
Collapse
|
11
|
Li D, Hu C, Su J, Du S, Zhang Y, Ni W, Ren L, Hao Y, Feng Y, Jin C, Wang S, Dai X, Wang Z, Zhu B, Xiao J, Shao Y. Function and structure of broadly neutralizing antibodies against SARS-CoV-2 Omicron variants isolated from prototype strain infected convalescents. J Transl Med 2025; 23:212. [PMID: 39985112 PMCID: PMC11844185 DOI: 10.1186/s12967-025-06162-6] [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: 09/13/2024] [Accepted: 01/22/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The ongoing emergence of evolving SARS-CoV-2 variants poses great threaten to the efficacy of authorized monoclonal antibody-based passive immunization or treatments. Developing potent broadly neutralizing antibodies (bNabs) against SARS-CoV-2 and elucidating their potential evolutionary pathways are essential for battling the coronavirus disease 2019 (COVID-19) pandemic. METHODS Broadly neutralizing antibodies were isolated using single cell sorting from three COVID-19 convalescents infected with prototype SARS-CoV-2 strain. Their neutralizing activity against diverse SARS-CoV-2 strains were tested in vitro and in vivo, respectively. The structures of antibody-antigen complexes were resolved using crystallization or Cryo-EM method. Antibodyomics analyses were performed using the non-bias deep sequencing results of BCR repertoires. RESULTS We obtained a series of RBD-specific monoclonal antibodies with highly neutralizing potency against a variety of pseudotyped and live SARS-CoV-2 variants, including five global VOCs and some Omicron subtypes such as BA.1, BA.2, BA.4/5, BF.7, and XBB. 2YYQH9 and LQLD6HL antibody cocktail also displayed good therapeutic and prophylactic efficacy in an XBB.1.16 infected hamster animal model. Cryo-EM and crystal structural analyses revealed that broadly neutralizing antibodies directly blocked the binding of ACE2 by almost covering the entire receptor binding motif (RBM) and largely avoided mutated RBD residues in the VOCs, demonstrating their broad and potent neutralizing activity. In addition, antibodyomics assays indicate that the germline frequencies of RBD-specific antibodies increase after an inactivated vaccine immunization. Moreover, the CDR3 frequencies of Vκ/λ presenting high amino acid identity with the broadly neutralizing antibodies were higher than those of VH. CONCLUSIONS These data suggest that current identified broadly neutralizing antibodies could serve as promising drug candidates for COVID-19 and can be used for reverse vaccine design against future pandemics.
Collapse
Affiliation(s)
- Dan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Caiqin Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Junwei Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Shuo Du
- Changping Laboratory, Beijing, 102206, China
| | - Ying Zhang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100091, China
| | - Wanqi Ni
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Li Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yanling Hao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yi Feng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Changzhong Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Shuo Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Xinxian Dai
- National Vaccine and Serum Institute, Beijing, 101111, China
- China National Biotec Group Company Limited, Beijing, 100024, China
| | - Zheng Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| | - Biao Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| | - Junyu Xiao
- Changping Laboratory, Beijing, 102206, China.
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100091, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100091, China.
| | - Yiming Shao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/ STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Changping Laboratory, Beijing, 102206, China.
| |
Collapse
|
12
|
Deraje P, Kitchens J, Coop G, Osmond MM. The promise and challenge of spatial inference with the full ancestral recombination graph under Brownian motion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.10.588900. [PMID: 40027772 PMCID: PMC11870416 DOI: 10.1101/2024.04.10.588900] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Spatial patterns of genetic relatedness among samples reflect the past movements of their ancestors. Our ability to untangle this history has the potential to improve dramatically given that we can now infer the ultimate description of genetic relatedness, the ancestral recombination graph (ARG). By extending spatial theory previously applied to trees, we generalize the common model of Brownian motion to full ARGs, thereby accounting for correlations in trees along a chromosome while efficiently computing likelihood-based estimates of dispersal rate and genetic ancestor locations, with associated uncertainties. We evaluate this model's ability to reconstruct spatial histories using individual-based simulations and unfortunately find a clear bias in the estimates of dispersal rate and ancestor locations. We investigate the causes of this bias, pinpointing a discrepancy between the model and the true spatial process at recombination events. This highlights a key hurdle in extending the ubiquitous and analytically-tractable model of Brownian motion from trees to ARGs, which otherwise has the potential to provide an efficient method for spatial inference, with uncertainties, using all the information available in the full ARG.
Collapse
Affiliation(s)
- Puneeth Deraje
- Department of Ecology & Evolutionary Biology, University of Toronto
| | - James Kitchens
- Department of Evolution & Ecology and Center for Population Biology, University of California - Davis
| | - Graham Coop
- Department of Evolution & Ecology and Center for Population Biology, University of California - Davis
| | | |
Collapse
|
13
|
Kamelian K, Sievers B, Chen-Xu M, Turner S, Cheng MTK, Altaf M, Kemp SA, Abdullahi A, Csiba K, Collier DA, Mlcochova P, Meng B, Jones RB, Smith D, Bradley J, Smith KGC, Doffinger R, Smith RM, Gupta RK. Humoral responses to SARS-CoV-2 vaccine in vasculitis-related immune suppression. SCIENCE ADVANCES 2025; 11:eadq3342. [PMID: 39937891 PMCID: PMC11817922 DOI: 10.1126/sciadv.adq3342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025]
Abstract
Immune suppression poses a challenge to vaccine immunogenicity. We show that serum antibody neutralization against SARS-CoV-2 Omicron descendants was largely absent post-doses 1 and 2 in individuals with vasculitis treated with rituximab. Detectable and increasing neutralizing titers were observed post-doses 3 and 4, except for XBB. Rituximab in vasculitis exacerbates neutralization deficits over standard immunosuppressive therapy, although impairment resolves over time since dosing. We observed discordance between detectable IgG binding and neutralizing activity specifically in the context of rituximab use, with high proportions of individuals showing reasonable IgG titer but no neutralization. ADCC response was more frequently detectable compared to neutralization in the context of rituximab, indicating that a notable proportion of binding antibodies are non-neutralizing. Therefore, use of rituximab is associated with severe impairment in neutralization against Omicron descendants despite repeated vaccinations, with better preservation of non-neutralizing antibody activity.
Collapse
Affiliation(s)
- Kimia Kamelian
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Benjamin Sievers
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Michael Chen-Xu
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, Cambridgeshire, UK
| | - Sam Turner
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Mark Tsz Kin Cheng
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Mazharul Altaf
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Steven A. Kemp
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Kata Csiba
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
| | - Dami A. Collier
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Bo Meng
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
| | - Rachel B. Jones
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, Cambridgeshire, UK
| | | | - Derek Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - John Bradley
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Department of Renal Medicine, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Kenneth G. C. Smith
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia
- University of Melbourne, Melbourne, VIC 3010, Australia
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrooke’s Hospital, Cambridge, UK
| | - Rona M. Smith
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, Cambridgeshire, UK
- Department of Renal Medicine, Addenbrooke’s Hospital, Cambridge, Cambridgeshire, UK
| | - Ravindra K. Gupta
- School of Clinical Medicine, Department of Medicine, University of Cambridge, Cambridge, Cambridgeshire, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, Cambridgeshire, UK
- Africa Health Research Institute, Durban, South Africa
| |
Collapse
|
14
|
Yousefbeigi S, Marsusi F. Structural insights into ACE2 interactions and immune activation of SARS-CoV-2 and its variants: an in-silico study. J Biomol Struct Dyn 2025; 43:665-678. [PMID: 37982275 DOI: 10.1080/07391102.2023.2283158] [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: 10/15/2022] [Accepted: 11/08/2023] [Indexed: 11/21/2023]
Abstract
The initial interaction between COVID-19 and the human body involves the receptor-binding domain (RBD) of the viral spike protein with the angiotensin-converting enzyme 2 (ACE2) receptor. Likewise, the spike protein can engage with immune-related proteins, such as toll-like receptors (TLRs) and pulmonary surfactant proteins A (SP-A) and D (SP-D), thereby triggering immune responses. In this study, we utilize computational methods to investigate the interactions between the spike protein and TLRs (specifically TLR2 and TLR4), as well as (SP-A) and (SP-D). The study is conducted on four variants of concern (VOC) to differentiate and identify common virus behaviours. An assessment of the structural stability of various variants indicates slight changes attributed to mutations, yet overall structural integrity remains preserved. Our findings reveal the spike protein's ability to bind with TLR4 and TLR2, prompting immune activation. In addition, our in-silico results reveal almost similar docking scores and therefore affinity for both ACE2-spike and TLR4-spike complexes. We demonstrate that even minor changes due to mutations in all variants, surfactant A and D proteins can function as inhibitors against the spike in all variants, hindering the ACE2-RBD interaction.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sarina Yousefbeigi
- Department of Physics and Energy Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Farah Marsusi
- Department of Physics and Energy Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
15
|
Rouzine IM. Evolutionary Mechanisms of the Emergence of the Variants of Concern of SARS-CoV-2. Viruses 2025; 17:197. [PMID: 40006952 PMCID: PMC11861269 DOI: 10.3390/v17020197] [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: 01/07/2025] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The evolutionary origin of the variants of concern (VOCs) of SARS-CoV-2, characterized by a large number of new substitutions and strong changes in virulence and transmission rate, is intensely debated. The leading explanation in the literature is a chronic infection in immunocompromised individuals, where the virus evolves before returning into the main population. The present article reviews less-investigated hypotheses of VOC emergence with transmission between acutely infected hosts, with a focus on the mathematical models of stochastic evolution that have proved to be useful for other viruses, such as HIV and influenza virus. The central message is that understanding the acting factors of VOC evolution requires the framework of stochastic multi-locus evolution models, and that alternative hypotheses can be effectively verified by fitting results of computer simulation to empirical data.
Collapse
Affiliation(s)
- Igor M Rouzine
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg 194223, Russia
| |
Collapse
|
16
|
Savulescu C, Prats-Uribe A, Brolin K, Uusküla A, Bergin C, Fleming C, Zvirbulis V, Zavadska D, Szułdrzyński K, Gaio V, Popescu CP, Craiu M, Cisneros M, Latorre-Millán M, Lohur L, McGrath J, Ferguson L, Abolina I, Gravele D, Machado A, Florescu SA, Lazar M, Subirats P, Clusa Cuesta L, Sui J, Kenny C, Krievins D, Barzdina EA, Melo A, Kosa AG, Miron VD, Muñoz-Almagro C, Milagro AM, Bacci S, Kramarz P, Nardone A. Relative effectiveness of the second booster COVID-19 vaccines against laboratory confirmed SARS-CoV-2 infection in healthcare workers: VEBIS HCW VE cohort study (1 October 2022-2 May 2023). Vaccine 2025; 45:126615. [PMID: 39724693 PMCID: PMC11790529 DOI: 10.1016/j.vaccine.2024.126615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Repeated COVID-19 booster vaccination was recommended in healthcare workers (HCWs) to maintain protection. We measured the relative vaccine effectiveness (rVE) of the second booster dose of COVID-19 vaccine compared to the first booster, against laboratory-confirmed SARS-CoV-2 infection in HCWs. METHODS In a prospective cohort study among HCWs from 12 European hospitals, we collected nasopharyngeal or saliva samples at enrolment and during weekly/fortnightly follow-up between October 2022 and May 2023. We estimated rVE of the second versus first COVID-19 vaccine booster dose against SARS-CoV-2 infection, overall, by time since second booster and restricted to the bivalent vaccines only. Using Cox regression, we calculated the rVE as (1-hazard ratio)*100, adjusting for hospital, age, sex, prior SARS-CoV-2 infection and at least one underlying condition. RESULTS Among the 979 included HCWs eligible for a second booster vaccination, 392 (40 %) received it and 192 (20 %) presented an infection during the study period. The rVE of the second versus first booster dose was -5 % (95 %CI: -46; 25) overall, 3 % (-46; 36) in the 7-89 days after receiving the second booster dose. The rVE was 11 % (-43; 45) when restricted to the use of bivalent vaccines only. CONCLUSION The bivalent COVID-19 could have reduced the risk of SARS-CoV-2 infection among HCWs by 11 %. However, we note the limitation of imprecise rVE estimates due to the proportion of monovalent vaccine used in the study, the small sample size and the study being conducted during the predominant circulation of XBB.1.5 sub-lineage. COVID-19 vaccine effectiveness studies in HCWs can provide important evidence to inform the optimal timing and the use of updated COVID-19 vaccines.
Collapse
Affiliation(s)
| | | | - Kim Brolin
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | - Anneli Uusküla
- Institute of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Colm Bergin
- Department of Genitourinary Medicine and Infectious Diseases (GUIDe), St. James's Hospital, Dublin, Ireland; Department of Clinical Medicine, Trinity College, Dublin, Ireland
| | - Catherine Fleming
- Galway University Hospital, Galway, Ireland; Department of Medicine, University of Galway, Dublin, Ireland
| | | | | | - Konstanty Szułdrzyński
- National Institute of Medicine of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Vânia Gaio
- Department of Epidemiology, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - Corneliu Petru Popescu
- Victor Babes Clinical Hospital of Infectious and Tropical Diseases, Bucharest, Romania; Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihai Craiu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; National Institute for Mother and Child Care Alessandrescu Rusescu, Bucharest, Romania
| | - Maria Cisneros
- Institut de Recerca Sant Joan de Deu. Hospital Sant Joan de Deu, Barcelona, Spain; Medicine Department, Universitat internacional de Catalunya, Barcelona, Spain
| | - Miriam Latorre-Millán
- Research Group on Difficult to Diagnose and Treat Infections, Miguel Servet University Hospital, IIS, Aragon, Zaragoza, Spain
| | | | - Jonathan McGrath
- Department of Genitourinary Medicine and Infectious Diseases (GUIDe), St. James's Hospital, Dublin, Ireland
| | | | - Ilze Abolina
- Pauls Stradins Clinical University Hospital, Riga, Latvia
| | | | - Ausenda Machado
- Department of Epidemiology, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - Simin Aysel Florescu
- Victor Babes Clinical Hospital of Infectious and Tropical Diseases, Bucharest, Romania; Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihaela Lazar
- Cantacuzino National Military-Medical Institute for Research and Development, Bucharest, Romania
| | - Pilar Subirats
- Department of Occupational Risk Prevention, Hospital Sant Joan de Deu, Barcelona, Spain
| | - Laura Clusa Cuesta
- Research Group on Difficult to Diagnose and Treat Infections, Miguel Servet University Hospital, IIS, Aragon, Zaragoza, Spain
| | - Jacklyn Sui
- Department of Genitourinary Medicine and Infectious Diseases (GUIDe), St. James's Hospital, Dublin, Ireland
| | | | | | | | - Aryse Melo
- Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge, Lisbon, Portugal
| | - Alma Gabriela Kosa
- Victor Babes Clinical Hospital of Infectious and Tropical Diseases, Bucharest, Romania
| | - Victor Daniel Miron
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; National Institute for Mother and Child Care Alessandrescu Rusescu, Bucharest, Romania
| | - Carmen Muñoz-Almagro
- Institut de Recerca Sant Joan de Deu. Hospital Sant Joan de Deu, Barcelona, Spain; Ciber of Epidemiology and Public Health CIBERESP, Madrid,Spain; Medicine Department, Universitat internacional de Catalunya, Barcelona, Spain
| | - Ana María Milagro
- Research Group on Difficult to Diagnose and Treat Infections, Miguel Servet University Hospital, IIS, Aragon, Zaragoza, Spain
| | - Sabrina Bacci
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | - Piotr Kramarz
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | | |
Collapse
|
17
|
Otoguro T, Wagatsuma K, Hino T, Ichikawa Y, Purnama TB, Sun Y, Li J, Chon I, Watanabe H, Saito R. Antibody Responses to mRNA COVID-19 Vaccine Among Healthcare Workers in Outpatient Clinics in Japan. Vaccines (Basel) 2025; 13:90. [PMID: 39852868 PMCID: PMC11769486 DOI: 10.3390/vaccines13010090] [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: 12/22/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND This study aimed to assess the antibody response to SARS-CoV-2 vaccines among healthcare workers (HCWs) from multiple outpatient clinics in Japan, examining the effects of baseline characteristics (e.g., sex, age, underlying condition, smoking history, occupation) and prior infections. METHODS A total of 101 HCWs provided serum at four time points between October 2020 and July 2023. HCWs received two to six doses of mRNA vaccine (BNT162b2 or mRNA-1273). Anti-nucleocapsid (N) and anti-spike (S) IgG antibodies against the ancestral Wuhan strain were measured using the Abbott Architect™ SARS-CoV-2 IgG assay. Univariate and regression analysis evaluated factors such as past infections, age, sex, smoking, underlying condition, and occupation. RESULTS After four to six doses, the median anti-S IgG titer in uninfected HCWs was 1807.30 BAU/mL, compared to 1899.89 BAU/mL in HCWs with prior infections. The median anti-N IgG titer was 0.10 index S/C in uninfected HCWs and 0.39 index S/C in infected HCWs. HCWs with prior infection had anti-S IgG titers 1.1 to 5.8 times higher than those without. Univariate and multivariate analyses indicated infection and vaccination significantly increased anti-S and anti-N IgG titers. Age, sex, smoking history and occupation did not influence antibody titers while underlying conditions were associated with lower anti-N IgG titers. CONCLUSIONS Infection and vaccination were strongly associated with an increase in anti-S and anti-N IgG titers; however, the impact of hybrid immunity appeared to be limited and varied depending on the timing of the sampling. These findings provide valuable insights for developing personalized vaccination strategies and future vaccine development.
Collapse
Affiliation(s)
- Teruhime Otoguro
- Infectious Diseases Research Center of Niigata University in Myanmar, Niigata University, Niigata 950-8510, Japan; (H.W.); (R.S.)
| | - Keita Wagatsuma
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata 950-8510, Japan; (K.W.); (Y.I.); (T.B.P.); (Y.S.); (J.L.); (I.C.)
- Institute for Research Administration, Niigata University, Niigata 950-8510, Japan
| | - Toshiharu Hino
- Hino Pediatric Internal Medicine Clinic, Nishinomiya 662-0927, Japan;
| | | | - Yusuke Ichikawa
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata 950-8510, Japan; (K.W.); (Y.I.); (T.B.P.); (Y.S.); (J.L.); (I.C.)
| | - Tri Bayu Purnama
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata 950-8510, Japan; (K.W.); (Y.I.); (T.B.P.); (Y.S.); (J.L.); (I.C.)
- Faculty of Public Health, Universitas Islam Negeri Sumatera Utara, Medan 20371, Indonesia
| | - Yuyang Sun
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata 950-8510, Japan; (K.W.); (Y.I.); (T.B.P.); (Y.S.); (J.L.); (I.C.)
| | - Jiaming Li
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata 950-8510, Japan; (K.W.); (Y.I.); (T.B.P.); (Y.S.); (J.L.); (I.C.)
| | - Irina Chon
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata 950-8510, Japan; (K.W.); (Y.I.); (T.B.P.); (Y.S.); (J.L.); (I.C.)
| | - Hisami Watanabe
- Infectious Diseases Research Center of Niigata University in Myanmar, Niigata University, Niigata 950-8510, Japan; (H.W.); (R.S.)
| | - Reiko Saito
- Infectious Diseases Research Center of Niigata University in Myanmar, Niigata University, Niigata 950-8510, Japan; (H.W.); (R.S.)
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata 950-8510, Japan; (K.W.); (Y.I.); (T.B.P.); (Y.S.); (J.L.); (I.C.)
| |
Collapse
|
18
|
Suntronwong N, Kanokudom S, Duangchinda T, Chantima W, Pakchotanon P, Klinfueng S, Puenpa J, Thatsanathorn T, Wanlapakorn N, Poovorawan Y. Neutralization of omicron subvariants and antigenic cartography following multiple COVID 19 vaccinations and repeated omicron non JN.1 or JN.1 infections. Sci Rep 2025; 15:1454. [PMID: 39789099 PMCID: PMC11718010 DOI: 10.1038/s41598-024-84138-0] [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: 10/15/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
The ongoing emergence of SARS-CoV-2 variants, combined with antigen exposures from different waves and vaccinations, poses challenges in updating COVID-19 vaccine antigens. We collected 206 sera from individuals with vaccination-only, hybrid immunity, and single or repeated omicron post-vaccination infections (PVIs), including non-JN.1 and JN.1, and evaluated neutralization against omicron BA.5, BA.2.75, BQ.1.1, XBB.1.16, XBB.1.5, and JN.1. Neutralizing antibodies exhibited a narrow breadth against BA.5 and BA.2.75 and failed to neutralize BQ.1.1 and XBB lineages after three to five doses of the ancestral monovalent vaccine. Hybrid immunity elicited higher neutralizing titers than vaccination alone, but titers remained relatively low. A single omicron PVI elicited lower neutralization titers to all variants compared to wild-type (WT), indicating immunological imprinting. Repeated omicron PVIs, particularly JN.1, slightly mitigated these effects by increasing broad neutralization responses to all variants, though not significantly. Antigenic mapping demonstrated that XBB lineages and JN.1 are antigenically distant from WT and also evaded antibodies induced by earlier omicron variants (BA.1-5) PVIs. However, repeated JN.1 PVIs shortened this antigenic distance, indicating broader neutralization across omicron variants. These findings highlight SARS-CoV-2 immunity following various antigen boosts and the impact of repeated omicron JN.1 exposure on broad immunity, informing future COVID-19 vaccination strategies.
Collapse
Affiliation(s)
- Nungruthai Suntronwong
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sitthichai Kanokudom
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Osteoarthritis and Musculoskeleton, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Thaneeya Duangchinda
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Development Agency, NSTDA, Pathum Thani, 12120, Thailand
| | - Warangkana Chantima
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pattarakul Pakchotanon
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Development Agency, NSTDA, Pathum Thani, 12120, Thailand
| | - Sirapa Klinfueng
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Jiratchaya Puenpa
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thaksaporn Thatsanathorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nasamon Wanlapakorn
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- The Royal Society of Thailand (FRS(T)), Sanam Sueapa, Dusit, Bangkok, 10330, Thailand.
| |
Collapse
|
19
|
Arantes I, Ito K, Gomes M, de Carvalho FC, Ferreira de Almeida WA, Khouri R, Miyajima F, Wallau GL, Naveca FG, Pereira EC, Mendonça Siqueira M, Resende PC, Bello G. Rapid spread of the SARS-CoV-2 Omicron XDR lineage derived from recombination between XBB and BA.2.86 subvariants circulating in Brazil in late 2023. Microbiol Spectr 2025; 13:e0119324. [PMID: 39611827 PMCID: PMC11705947 DOI: 10.1128/spectrum.01193-24] [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: 05/21/2024] [Accepted: 09/21/2024] [Indexed: 11/30/2024] Open
Abstract
Recombination plays a crucial role in the evolution of SARS-CoV-2. The Omicron XBB* recombinant lineages are a noteworthy example, as they have been the dominant SARS-CoV-2 variant worldwide in the first half of 2023. Since November 2023, a new recombinant lineage between Omicron subvariants XBB and BA.2.86, designated XDR, has been detected mainly in Brazil. In this study, we reconstructed the spatiotemporal dynamics and estimated the absolute and relative transmissibility of the XDR lineage. The XDR lineage displayed a recombination breakpoint in the ORF1a-coding region, and the most closely related sequences to the 5' and 3' ends of the recombinant correspond to JD.1.1 and JN.1.1 lineages, respectively. The first XDR sequences were detected in November 2023 in the Northeastern Brazilian region, and their prevalence rapidly surged from <1% to 25% by February 2024. The Bayesian phylogeographic analysis supports that the XDR lineage likely emerged in the Northeastern Brazilian region around late October 2023 and rapidly disseminated within and outside Brazilian borders from mid-November onward. The median effective reproductive number of the XDR lineage in Brazil during the initial expansion phase was estimated to be around 1.5, and the average relative instantaneous reproduction numbers of XDR and JN* lineages were estimated to be 1.37 and 1.29 higher than that of co-circulating XBB* lineages. In summary, these findings support that the recombinant lineage XDR arose in the Northeastern Brazilian region in October 2023, shortly after the first detection of JN.1 sequences in the country. In Brazil, the XDR lineage exhibited a higher transmissibility level than its parental XBB.* lineages and is spreading at a rate similar to or slightly faster than the JN.1* lineages.IMPORTANCEThis study highlights the emergence and rapid dissemination of the recombinant SARS-CoV-2 XDR lineage, derived from the Omicron lineages JD.1.1 and JN.1.1. The XDR lineage exhibited equivalent transmissibility to its JN.1* parental lineages and quickly spread across Brazil in late 2023. The findings underscore the critical role of real-time genomic surveillance in detecting novel variants with higher transmission potential. By utilizing phylogenetic and epidemiological methods, this research provides important insights into the molecular dynamics of XDR, which could inform public health responses and vaccine composition updates. The study's significance lies in its ability to document the impact of recombination on viral evolution, offering valuable information to the field of virology and pandemic preparedness.
Collapse
Affiliation(s)
- Ighor Arantes
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Kimihito Ito
- International Institute for Zoonosis Control, Hokkaido University, Hokkaido, Japan
| | - Marcelo Gomes
- Grupo de Métodos Analíticos em Vigilância Epidemiológica, Fiocruz, Rio de Janeiro, Brazil
| | - Felipe Cotrim de Carvalho
- Departamento do Programa Nacional de Imunizações, Coordenação-Geral de Vigilância das doenças imunopreveníveis, Secretaria de Vigilância em saúde e ambiente, Brasília, Brazil
| | - Walquiria Aparecida Ferreira de Almeida
- Departamento do Programa Nacional de Imunizações, Coordenação-Geral de Vigilância das doenças imunopreveníveis, Secretaria de Vigilância em saúde e ambiente, Brasília, Brazil
| | | | | | - Gabriel Luz Wallau
- />Department of Arbovirology, Bernhard Nocht Institute for Tropical Medicine, WHO Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, National Reference Center for Tropical Infectious Diseases, Hamburg, Germany
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Pernambuco, Brazil
| | - Felipe Gomes Naveca
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Núcleo de Vigilância de Vírus Emergentes, Reemergentes ou Negligenciados, Laboratório de Ecologia de Doenças Transmissíveis na Amazônia, Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
| | - Elisa Cavalcante Pereira
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - COVID-19 Fiocruz Genomic Surveillance Network
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- International Institute for Zoonosis Control, Hokkaido University, Hokkaido, Japan
- Grupo de Métodos Analíticos em Vigilância Epidemiológica, Fiocruz, Rio de Janeiro, Brazil
- Departamento do Programa Nacional de Imunizações, Coordenação-Geral de Vigilância das doenças imunopreveníveis, Secretaria de Vigilância em saúde e ambiente, Brasília, Brazil
- Instituto Gonçalo Moniz, Fiocruz, Salvador, Brazil
- Fiocruz Ceará, Fortaleza, Brazil
- />Department of Arbovirology, Bernhard Nocht Institute for Tropical Medicine, WHO Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, National Reference Center for Tropical Infectious Diseases, Hamburg, Germany
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Pernambuco, Brazil
- Núcleo de Vigilância de Vírus Emergentes, Reemergentes ou Negligenciados, Laboratório de Ecologia de Doenças Transmissíveis na Amazônia, Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Marilda Mendonça Siqueira
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Paola Cristina Resende
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Gonzalo Bello
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Wang H, Peng Q, Dai X, Ying Z, Wu X, Liu X, Xu H, Li J, Shi L, Liu J, Wang Y, Zhao D, Huang Y, Yang L, Yang R, Yue G, Suo Y, Ye Q, Cao S, Li Y. A SARS-CoV-2 EG.5 mRNA vaccine induces a broad-spectrum immune response in mice. MedComm (Beijing) 2025; 6:e779. [PMID: 39760111 PMCID: PMC11695206 DOI: 10.1002/mco2.779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 01/07/2025] Open
Abstract
The emerging of emergent SARS-CoV-2 subvariants has reduced the protective efficacy of COVID-19 vaccines. Therefore, novel COVID-19 vaccines targeting these emergent variants are needed. We designed and prepared CoV072, an mRNA-based vaccine against SARS-CoV-2 Omicron (EG.5) and other emergent SARS-CoV-2 subvariants that encodes the EG.5 spike protein. Six-week-old female BALB/C mice were used to assess humoral and cellular immune responses and cross-reactive neutralizing activity against various SARS-CoV-2 subvariants. Meanwhile different immunization strategies and doses were performed to detect the immunogenicity of this mRNA vaccine. Our results show that two doses of 5 µg CoV072 or a single dose of 15 µg CoV072 both induced broad-spectrum cross-protection ability in mice. Compared with a single dose of 15 µg CoV072, two doses of 5 µg COV072 exhibited higher levels of pseudovirus neutralizing antibody (PNAb) and cross-reactive IgG responses to multiple variants. Moreover, higher levels of neutralizing antibody (NAb) against live XBB and EG.5 variants were also induced. Th1-biased cellular immune response was induced in all vaccination groups. The antigen design and immunization strategy of this study have reference significance for the research of the next generation of COVID-19 vaccine and other vaccines.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Qinhua Peng
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Xinxian Dai
- Etiology Laboratory,National Vaccine and Serum InstituteBeijingChina
| | - Zhifang Ying
- Division of Respiratory Virus VaccinesNational Institutes for Food and Drug ControlBeijingChina
| | - Xiaohong Wu
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Xinyu Liu
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Hongshan Xu
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Jia Li
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Leitai Shi
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Jingjing Liu
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Yunpeng Wang
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Danhua Zhao
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Yanqiu Huang
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Lihong Yang
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Ren Yang
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Guangzhi Yue
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Yue Suo
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Qiang Ye
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Shouchun Cao
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| | - Yuhua Li
- Department of Arboviral VaccineNational Institutes for Food and Drug ControlBeijingChina
| |
Collapse
|
21
|
Mears HV, Young GR, Sanderson T, Harvey R, Barrett-Rodger J, Penn R, Cowton V, Furnon W, De Lorenzo G, Crawford M, Snell DM, Fowler AS, Chakrabarti AM, Hussain S, Gilbride C, Emmott E, Finsterbusch K, Luptak J, Peacock TP, Nicod J, Patel AH, Palmarini M, Wall E, Williams B, Gandhi S, Swanton C, Bauer DLV. Emergence of SARS-CoV-2 subgenomic RNAs that enhance viral fitness and immune evasion. PLoS Biol 2025; 23:e3002982. [PMID: 39836705 PMCID: PMC11774490 DOI: 10.1371/journal.pbio.3002982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/28/2025] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Coronaviruses express their structural and accessory genes via a set of subgenomic RNAs, whose synthesis is directed by transcription regulatory sequences (TRSs) in the 5' genomic leader and upstream of each body open reading frame. In SARS-CoV-2, the TRS has the consensus AAACGAAC; upon searching for emergence of this motif in the global SARS-CoV-2 sequences, we find that it evolves frequently, especially in the 3' end of the genome. We show well-supported examples upstream of the Spike gene-within the nsp16 coding region of ORF1b-which is expressed during human infection, and upstream of the canonical Envelope gene TRS, both of which have evolved convergently in multiple lineages. The most frequent neo-TRS is within the coding region of the Nucleocapsid gene, and is present in virtually all viruses from the B.1.1 lineage, including the variants of concern Alpha, Gamma, Omicron and descendants thereof. Here, we demonstrate that this TRS leads to the expression of a novel subgenomic mRNA encoding a truncated C-terminal portion of Nucleocapsid, which is an antagonist of type I interferon production and contributes to viral fitness during infection. We observe distinct phenotypes when the Nucleocapsid coding sequence is mutated compared to when the TRS alone is ablated. Our findings demonstrate that SARS-CoV-2 is undergoing evolutionary changes at the functional RNA level in addition to the amino acid level.
Collapse
Affiliation(s)
- Harriet V. Mears
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
| | - George R. Young
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
- Bioinformatics and Biostatistics STP, The Francis Crick Institute, London, United Kingdom
| | - Theo Sanderson
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ruth Harvey
- Worldwide Influenza Centre, The Francis Crick Institute, London, United Kingdom
| | - Jamie Barrett-Rodger
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Rebecca Penn
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Vanessa Cowton
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Wilhelm Furnon
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Giuditta De Lorenzo
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | | | - Daniel M. Snell
- Genomics STP, The Francis Crick Institute, London, United Kingdom
| | - Ashley S. Fowler
- Genomics STP, The Francis Crick Institute, London, United Kingdom
| | - Anob M. Chakrabarti
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
- UCL Respiratory, Division of Medicine, UCL, London, United Kingdom
| | - Saira Hussain
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ciarán Gilbride
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Edward Emmott
- Centre for Proteome Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Katja Finsterbusch
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Jakub Luptak
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Thomas P. Peacock
- Department of Infectious Disease, St Mary’s Hospital, Imperial College London, London, United Kingdom
| | - Jérôme Nicod
- Genomics STP, The Francis Crick Institute, London, United Kingdom
| | - Arvind H. Patel
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
- Genotype-to-Phenotype (G2P-UK) National Virology Consortium, London, United Kingdom
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
- Genotype-to-Phenotype (G2P-UK) National Virology Consortium, London, United Kingdom
| | - Emma Wall
- Crick/UCLH Legacy Study, The Francis Crick Institute, London, United Kingdom
- University College London and National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, United Kingdom
| | - Bryan Williams
- University College London and National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, London, United Kingdom
| | - Sonia Gandhi
- Neurodegeneration Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - David L. V. Bauer
- RNA Virus Replication Laboratory, The Francis Crick Institute, London, United Kingdom
- Genotype-to-Phenotype (G2P-UK) National Virology Consortium, London, United Kingdom
| |
Collapse
|
22
|
Angius F, Puxeddu S, Zaimi S, Canton S, Nematollahzadeh S, Pibiri A, Delogu I, Alvisi G, Moi ML, Manzin A. SARS-CoV-2 Evolution: Implications for Diagnosis, Treatment, Vaccine Effectiveness and Development. Vaccines (Basel) 2024; 13:17. [PMID: 39852796 PMCID: PMC11769326 DOI: 10.3390/vaccines13010017] [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: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, driven by the rapid evolution of the SARS-CoV-2 virus, presents ongoing challenges to global public health. SARS-CoV-2 is characterized by rapidly evolving mutations, especially in (but not limited to) the spike protein, complicating predictions about its evolutionary trajectory. These mutations have significantly affected transmissibility, immune evasion, and vaccine efficacy, leading to multiple pandemic waves with over half a billion cases and seven million deaths globally. Despite several strategies, from rapid vaccine development and administration to the design and availability of antivirals, including monoclonal antibodies, already having been employed, the persistent circulation of the virus and the emergence of new variants continue to result in high case numbers and fatalities. In the past four years, immense research efforts have contributed much to our understanding of the viral pathogenesis mechanism, the COVID-19 syndrome, and the host-microbe interactions, leading to the development of effective vaccines, diagnostic tools, and treatments. The focus of this review is to provide a comprehensive analysis of the functional impact of mutations on diagnosis, treatments, and vaccine effectiveness. We further discuss vaccine safety in pregnancy and the implications of hybrid immunity on long-term protection against infection, as well as the latest developments on a pan-coronavirus vaccine and nasal formulations, emphasizing the need for continued surveillance, research, and adaptive public health strategies in response to the ongoing SARS-CoV-2 evolution race.
Collapse
Affiliation(s)
- Fabrizio Angius
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvia Puxeddu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvio Zaimi
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Serena Canton
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Sepehr Nematollahzadeh
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Andrea Pibiri
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Ilenia Delogu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| |
Collapse
|
23
|
Sadhu S, Goswami S, Khatri R, Lohiya B, Singh V, Yadav R, Das V, Tripathy MR, Dwivedi P, Srivastava M, Mani S, Asthana S, Samal S, Awasthi A. Berbamine prevents SARS-CoV-2 entry and transmission. iScience 2024; 27:111347. [PMID: 39640591 PMCID: PMC11618033 DOI: 10.1016/j.isci.2024.111347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/17/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Effective antiviral drugs are essential to combat COVID-19 and future pandemics. Although many compounds show antiviral in vitro activity, only a few retain effectiveness in vivo against SARS-CoV-2. Here, we show that berbamine (Berb) is effective against SARS-CoV, MER-CoV, SARS-CoV-2 and its variants, including the XBB.1.16 variant. In hACE2.Tg mice, Berb suppresses SARS-CoV-2 replication through two distinct mechanisms: inhibiting spike-mediated viral entry and enhancing antiviral gene expression during infection. The administration of Berb, in combination with remdesivir (RDV), clofazimine (Clof) and fangchinoline (Fcn), nearly eliminated viral load and promoted recovery from acute SARS-CoV-2 infection and its variants. Co-housed mice in direct contact with either pre-treated or untreated infected mice exhibited negligible viral loads, reduced lung pathology, and decreased viral shedding, suggesting that Berb may effectively hinder virus transmission. This broad-spectrum activity positions Berb as a promising preventive or therapeutic option against betacoronaviruses.
Collapse
Affiliation(s)
- Srikanth Sadhu
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Sandeep Goswami
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Ritika Khatri
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Bharat Lohiya
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Virendra Singh
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Rahul Yadav
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Vinayaka Das
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Manas Ranjan Tripathy
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Prabhanjan Dwivedi
- Small Animal Facility, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Mitul Srivastava
- Non-communicable Disease Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Shailendra Mani
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Shailendra Asthana
- Non-communicable Disease Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Sweety Samal
- Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| | - Amit Awasthi
- Immuno-biology Lab, Infection and Immunology Centre, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
- Immunology-Core Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, Faridabad, Haryana 121001, India
| |
Collapse
|
24
|
Choga WT, Gobe I, Seru K, Maruapula D, Ndlovu NS, Zuze BJL, Motshosi P, Matsuru T, Sabone P, Yu X, Blackard JT, San JE, Makhema J, Gaseitsiwe S, Moyo S. Genomic epidemiology and immune escape of SARS-CoV-2 recombinant strains circulating in Botswana. IJID REGIONS 2024; 13:100484. [PMID: 39670194 PMCID: PMC11636131 DOI: 10.1016/j.ijregi.2024.100484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 12/14/2024]
Abstract
Objectives We characterized the molecular and mutational landscape of SARS-CoV-2 recombinant strains in Botswana. Methods We performed genomic, phylogenetic, and immunoinformatic analyses of 5254 near-complete genomes from 2020 to 2023. We assessed the presence of mutations of interested (MutOI) that may be associated with immune escape in silico. Results We observed a few recombinant strains in Botswana, with the majority being descendants of Omicron (XBB*), except for XV and XM. Most recombinant sequences corresponded to transmission clusters. Most recombination events occurred within the receptor-binding domain (RDB) of the spike (S) protein. We identified 16 MutOI among different proteins, with the majority occurring at a very low global prevalence (<4.8 × 10-⁵). We also observed S:Q474K, a MutOI in the RBD, that was predicted to escape human leukocyte antigen class I-mediated immune responses. Molecular surveillance is vital to inform early detection and response to potential variants with heightened immune and vaccine breakthrough properties. Conclusions These results underscore the need for continued molecular surveillance to map the evolutionary landscape of SARS-CoV-2.
Collapse
Affiliation(s)
- Wonderful T. Choga
- Botswana Harvard Health Partnership, Gaborone, Botswana
- School of Allied Health Sciences, Faculty of Health Sciences, Gaborone, Botswana
| | - Irene Gobe
- School of Allied Health Sciences, Faculty of Health Sciences, Gaborone, Botswana
| | | | | | | | | | | | - Teko Matsuru
- Botswana Harvard Health Partnership, Gaborone, Botswana
| | - Phenyo Sabone
- Botswana Harvard Health Partnership, Gaborone, Botswana
| | - Xiaoyu Yu
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, Scotland, UK
| | | | - James E. San
- Duke Human Vaccine Institute (DHVI), Durham, USA
- University of KwaZulu-Natal (UKZN), Durban, South Africa
| | | | - Simani Gaseitsiwe
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Sikhulile Moyo
- Botswana Harvard Health Partnership, Gaborone, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, USA
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
25
|
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
|
26
|
Kuwata T, Kaku Y, Biswas S, Matsumoto K, Shimizu M, Kawanami Y, Uraki R, Okazaki K, Minami R, Nagasaki Y, Nagashima M, Yoshida I, Sadamasu K, Yoshimura K, Ito M, Kiso M, Yamayoshi S, Imai M, Ikeda T, Sato K, Toyoda M, Ueno T, Inoue T, Tanaka Y, Kimura KT, Hashiguchi T, Sugita Y, Noda T, Morioka H, Kawaoka Y, Matsushita S. Induction of IGHV3-53 public antibodies with broadly neutralising activity against SARS-CoV-2 including Omicron subvariants in a Delta breakthrough infection case. EBioMedicine 2024; 110:105439. [PMID: 39488016 PMCID: PMC11565539 DOI: 10.1016/j.ebiom.2024.105439] [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: 02/13/2024] [Revised: 10/08/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Emergence of SARS-CoV-2 variants that escape neutralising antibodies hampers the development of vaccines and therapeutic antibodies against SARS-CoV-2. IGHV3-53/3-66-derived public antibodies, which are generally specific to the prototype virus and are frequently induced in infected or vaccinated individuals, show minimal affinity maturation and high potency against prototype SARS-CoV-2. METHODS Monoclonal antibodies isolated from a Delta breakthrough infection case were analysed for cross-neutralising activities against SARS-CoV-2 variants. The broadly neutralising antibody K4-66 was further analysed in a hamster model, and the effect of somatic hypermutations was assessed using the inferred germline precursor. FINDINGS Antibodies derived from IGHV3-53/3-66 showed broader neutralising activity than antibodies derived from IGHV1-69 and other IGHV genes. IGHV3-53/3-66 antibodies neutralised the Delta variant better than the IGHV1-69 antibodies, suggesting that the IGHV3-53/3-66 antibodies were further maturated by Delta breakthrough infection. One IGHV3-53/3-66 antibody, K4-66, neutralised all Omicron subvariants tested, including EG.5.1, BA.2.86, and JN.1, and decreased the viral load in the lungs of hamsters infected with Omicron subvariant XBB.1.5. The importance of somatic hypermutations was demonstrated by the loss of neutralising activity of the inferred germline precursor of K4-66 against Beta and Omicron variants. INTERPRETATION Broadly neutralising IGHV3-53/3-66 antibodies have potential as a target for the development of effective vaccines and therapeutic antibodies against newly emerging SARS-CoV-2 variants. FUNDING This work was supported by grants from AMED (JP23ym0126048, JP22ym0126048, JP21ym0126048, JP23wm0125002, JP233fa627001, JP223fa627009, JP24jf0126002, and JP22fk0108572), and the JSPS (JP21H02970, JK23K20041, and JPJSCCA20240006).
Collapse
Affiliation(s)
- Takeo Kuwata
- Collaborative Research Program with the Chemo-Sero-Therapeutic Research Institute for Anti-viral Agents and Hematological Diseases, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| | - Yu Kaku
- Collaborative Research Program with the Chemo-Sero-Therapeutic Research Institute for Anti-viral Agents and Hematological Diseases, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan; Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shashwata Biswas
- Collaborative Research Program with the Chemo-Sero-Therapeutic Research Institute for Anti-viral Agents and Hematological Diseases, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kaho Matsumoto
- Collaborative Research Program with the Chemo-Sero-Therapeutic Research Institute for Anti-viral Agents and Hematological Diseases, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Mikiko Shimizu
- Collaborative Research Program with the Chemo-Sero-Therapeutic Research Institute for Anti-viral Agents and Hematological Diseases, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Yoko Kawanami
- Collaborative Research Program with the Chemo-Sero-Therapeutic Research Institute for Anti-viral Agents and Hematological Diseases, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Ryuta Uraki
- Division of Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kyo Okazaki
- Department of Analytical and Biophysical Chemistry, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rumi Minami
- Internal Medicine, Clinical Research Institute, NHO Kyushu Medical Center, Fukuoka, Japan
| | - Yoji Nagasaki
- Internal Medicine, Clinical Research Institute, NHO Kyushu Medical Center, Fukuoka, Japan
| | - Mami Nagashima
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | - Isao Yoshida
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | - Kenji Sadamasu
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | | | - Mutsumi Ito
- Division of Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Maki Kiso
- Division of Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Yamayoshi
- Division of Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaki Imai
- Division of Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mako Toyoda
- Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Takamasa Ueno
- Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Takako Inoue
- Department of Clinical Laboratory Medicine, Nagoya City University Hospital, Nagoya, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kanako Tarakado Kimura
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yukihiko Sugita
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takeshi Noda
- Laboratory of Ultrastructural Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroshi Morioka
- Department of Analytical and Biophysical Chemistry, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuzo Matsushita
- Collaborative Research Program with the Chemo-Sero-Therapeutic Research Institute for Anti-viral Agents and Hematological Diseases, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
27
|
Gong YN, Kuo NY, Yeh TS, Shih SR, Chen GW. Genomic Surveillance of SARS-CoV-2 in Taiwan: A Perspective on Evolutionary Data Interpretation and Sequencing Issues. Biomed J 2024:100820. [PMID: 39608568 DOI: 10.1016/j.bj.2024.100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/26/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
This review presents a comprehensive perspective on the genomic surveillance of SARS-CoV-2 in Taiwan, with a focus on next-generation sequencing and phylogenetic interpretation. This article aimed to explore how Taiwan has utilized genomic sequencing technologies and surveillance to monitor and mitigate the spread of COVID-19. We examined databases and sources of genomic sequences and highlighted the role of data science methodologies in the explanation and analyses of evolutionary data. This review addressed the challenges and limitations inherent in genomic surveillance, such as concerns regarding data quality and the necessity for interdisciplinary expertise for accurate data interpretation. Special attention was given to the unique challenges faced by Taiwan, including its high population density and major transit destination for international travelers. We underscored the far-reaching implications of genomic surveillance data for public health policy, particularly in influencing decisions regarding travel restrictions, vaccine administration, and public health decision-making. Studies were examined to demonstrate the effectiveness of using genomic data to implement public health measures. Future research should prioritize the integration of methodologies and technologies in evolutionary data science, particularly focusing on phylodynamic analytics. This integration is crucial to enhance the precision and applicability of genomic data. Overall, we have provided an overview of the significance of genomic surveillance in tracking SARS-CoV-2 variants globally and the pivotal role of data science methodologies in interpreting these data for effective public health interventions.
Collapse
Affiliation(s)
- Yu-Nong Gong
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; International Master Degree Program for Molecular Medicine in Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Nai-Yu Kuo
- Medical Education Department, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Syuan Yeh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Guang-Wu Chen
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Computer Science and Information Engineering, College of Engineering, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
28
|
Bendall EE, Dimcheff D, Papalambros L, Fitzsimmons WJ, Zhu Y, Schmitz J, Halasa N, Chappell J, Martin ET, Biddle JE, Smith-Jeffcoat SE, Rolfes MA, Mellis A, Talbot HK, Grijalva C, Lauring AS. In depth sequencing of a serially sampled household cohort reveals the within-host dynamics of Omicron SARS-CoV-2 and rare selection of novel spike variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624722. [PMID: 39605326 PMCID: PMC11601520 DOI: 10.1101/2024.11.21.624722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
SARS-CoV-2 has undergone repeated and rapid evolution to circumvent host immunity. However, outside of prolonged infections in immunocompromised hosts, within-host positive selection has rarely been detected. The low diversity within-hosts and strong genetic linkage among genomic sites make accurately detecting positive selection difficult. Longitudinal sampling is a powerful method for detecting selection that has seldom been used for SARS-CoV-2. Here we combine longitudinal sampling with replicate sequencing to increase the accuracy of and lower the threshold for variant calling. We sequenced 577 specimens from 105 individuals from a household cohort primarily during the BA.1/BA.2 variant period. There was extremely low diversity and a low rate of divergence. Specimens had 0-12 intrahost single nucleotide variants (iSNV) at >0.5% frequency, and the majority of the iSNV were at frequencies <2%. Within-host dynamics were dominated by genetic drift and purifying selection. Positive selection was rare but highly concentrated in spike. Two individuals with BA.1 infections had S:371F, a lineage defining substitution for BA.2. A Wright Fisher Approximate Bayesian Computational model identified positive selection at 14 loci with 7 in spike, including S:448 and S:339. We also detected significant genetic hitchhiking between synonymous changes and nonsynonymous iSNV under selection. The detectable immune-mediated selection may be caused by the relatively narrow antibody repertoire in individuals during the early Omicron phase of the SARS-CoV-2 pandemic. As both the virus and population immunity evolve, understanding the corresponding shifts in SARS-CoV-2 within-host dynamics will be important.
Collapse
Affiliation(s)
- Emily E. Bendall
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Derek Dimcheff
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Leigh Papalambros
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan Schmitz
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Natasha Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | - H. Keipp Talbot
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carlos Grijalva
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam S. Lauring
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
29
|
Choga WT, Gustani-Buss E, Tegally H, Maruapula D, Yu X, Moir M, Zuze BJL, James SE, Ndlovu NS, Seru K, Motshosi P, Blenkinsop A, Gobe I, Baxter C, Manasa J, Lockman S, Shapiro R, Makhema J, Wilkinson E, Blackard JT, Lemey P, Lessells RJ, Martin DP, de Oliveira T, Gaseitsiwe S, Moyo S. Emergence of Omicron FN.1 a descendent of BQ.1.1 in Botswana. Virus Evol 2024; 10:veae095. [PMID: 39720788 PMCID: PMC11666700 DOI: 10.1093/ve/veae095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 10/31/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Botswana, like the rest of the world, has been significantly impacted by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In December 2022, we detected a monophyletic cluster of genomes comprising a sublineage of the Omicron variant of concern (VOC) designated as B.1.1.529.5.3.1.1.1.1.1.1.74.1 (alias FN.1, clade 22E). These genomes were sourced from both epidemiologically linked and unlinked samples collected in three close locations within the district of Greater Gaborone. In this study, we assessed the worldwide prevalence of the FN.1 lineage, evaluated its mutational profile, and conducted a phylogeographic analysis to reveal its global dispersal dynamics. Among approximately 16 million publicly available SARS-CoV-2 sequences generated by 30 September 2023, only 87 were of the FN.1 lineage, including 22 from Botswana, 6 from South Africa, and 59 from the UK. The estimated time to the most recent common ancestor of the 87 FN.1 sequences was 22 October 2022 [95% highest posterior density: 2 September 2022-24 November 2022], with the earliest of the 22 Botswana sequences having been sampled on 7 December 2022. Discrete trait reconstruction of FN.1 identified Botswana as the most probable place of origin. The FN.1 lineage is derived from the BQ.1.1 lineage and carries two missense variants in the spike protein, S:K182E in NTD and S:T478R in RDB. Among the over 90 SARS-CoV-2 lineages circulating in Botswana between September 2020 and July 2023, FN.1 was most closely related to BQ.1.1.74 based on maximum likelihood phylogenetic inference, differing only by the S:K182E mutation found in FN.1. Given the early detection of numerous novel variants from Botswana and its neighbouring countries, our study underscores the necessity of continuous surveillance to monitor the emergence of potential VOCs, integrating molecular and spatial data to identify dissemination patterns enhancing preparedness efforts.
Collapse
Affiliation(s)
- Wonderful T Choga
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
- Faculty of Health Sciences, School of Allied Health Sciences, Gaborone, Private Bag UB 0022, Botswana
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Emanuele Gustani-Buss
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven 3000, Belgium
| | - Houriiyah Tegally
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Dorcas Maruapula
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
| | - Xiaoyu Yu
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh EH9 3FL, Scotland, UK
| | - Monika Moir
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Boitumelo J L Zuze
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
- Faculty of Health Sciences, School of Allied Health Sciences, Gaborone, Private Bag UB 0022, Botswana
| | - San Emmanuel James
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory. Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Nokuthula S Ndlovu
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
| | - Kedumetse Seru
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
| | - Patience Motshosi
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
| | - Alexandra Blenkinsop
- Department of Mathematics, Imperial College London, London, Westminster, SW7 2AZ, United Kingdom
| | - Irene Gobe
- Faculty of Health Sciences, School of Allied Health Sciences, Gaborone, Private Bag UB 0022, Botswana
| | - Cheryl Baxter
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Justen Manasa
- Faculty of Medicine and Health Sciences, Molecular Diagnostics and Investigative Sciences, University of Zimbabwe, Harare, P.O.Box MP167, Zimbabwe
| | - Shahin Lockman
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States
- Division of Infectious Diseases, Brigham & Women’s Hospital, Boston, MA 02115, United States
- Harvard Medical School, Boston, MA, 02115, United States
| | - Roger Shapiro
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States
| | - Joseph Makhema
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States
| | - Eduan Wilkinson
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Jason T Blackard
- University of Cincinnati College of Medicine, Cincinnati, OH 45267, United States
| | - Phillipe Lemey
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven 3000, Belgium
| | - Richard J Lessells
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory. Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Darren P Martin
- Division of Computational Biology, Department of Integrative Biomedial Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa
| | - Tulio de Oliveira
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory. Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
- Department of Global Health, University of Washington, Seattle, WA 98105, United States
| | - Simani Gaseitsiwe
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States
| | - Sikhulile Moyo
- Research Laboratory, Botswana Harvard Health Partnership, Gaborone, Private Bag BO 320, Botswana
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, United States
- School of Health Systems and Public Health, University of Pretoria, Pretoria 0002, South Africa
- Division of Medical Virology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town 7602, South Africa
| |
Collapse
|
30
|
Chewaskulyong B, Satjaritanun P, Ketpueak T, Suksombooncharoen T, Charoentum C, Nuchpong N, Tantraworasin A. Neutralizing antibodies and safety of a COVID-19 vaccine against SARS-CoV-2 wild-type and Omicron variants in solid cancer patients. PLoS One 2024; 19:e0310781. [PMID: 39509358 PMCID: PMC11542819 DOI: 10.1371/journal.pone.0310781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024] Open
Abstract
OBJECTIVE The aim of this study was to assess the seroconversion rate and percent inhibition of neutralizing antibodies against the wild-type and Omicron variants of SARS-CoV-2 in patients with solid cancer who received two COVID-19 vaccine doses by comparing chemotherapy and nonchemotherapy groups. METHODS This prospective cohort study enrolled 115 cancer patients from Maharaj Nakorn Chiang Mai Hospital, Sriphat Medical Center, Faculty of Medicine, Chiang Mai University, and Chiang Mai Klaimor Hospital, Chiang Mai, Thailand, between August 2021 and February 2022, with data from 91 patients who received two COVID-19 vaccine doses analyzed. Participants received vaccines as part of their personal vaccination programs, including various mRNA and non-mRNA vaccine combinations. Blood samples were collected at baseline, on day 28, and at 6 months post-second dose to assess neutralizing antibodies. The primary outcome was the seroconversion rate against the wild-type and Omicron variants on day 28. Secondary outcomes included seroconversion at 6 months, factors associated with seroconversion, and safety. RESULTS Among the participants, 45% were receiving chemotherapy. On day 28, seroconversion rates were 77% and 62% for the wild-type and Omicron variants, respectively. Chemotherapy did not significantly affect seroconversion rates (p = 0.789 for wild type, p = 0.597 for Omicron). The vaccine type administered was positively correlated with seroconversion, with an adjusted odds ratio (95% confidence interval) of 25.86 (1.39-478.06) for the wild type and 17.38 (3.65-82.66) for the Omicron variant with the primary heterologous vaccine regimen. Grades 1 and 2 adverse events were observed in 34.0% and 19.7% of participants, respectively. CONCLUSIONS Despite the lower seroconversion rate against the Omicron variant, no significant difference was observed between the chemotherapy and nonchemotherapy groups. COVID-19 vaccinations demonstrated good tolerability in this cohort. These findings highlight the importance of vaccine safety and immunogenicity in cancer patients and can inform tailored vaccination strategies for this vulnerable population.
Collapse
Affiliation(s)
- Busyamas Chewaskulyong
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattarapong Satjaritanun
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanika Ketpueak
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thatthan Suksombooncharoen
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaiyut Charoentum
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nuttaphoom Nuchpong
- Medical Oncology Outpatient Clinic, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai University, Chiang Mai, Thailand
| | - Apichat Tantraworasin
- Department of Surgery, General Thoracic Unit, Faculty of Medicine, and Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
31
|
Kongsomros S, Boonyarattanasoonthorn T, Phongphaew W, Kasorndorkbua C, Sunyakumthorn P, Im-Erbsin R, Lugo-Roman LA, Kongratanapasert T, Paha J, Manopwisedjaroen S, Kwankhao P, Supannapan K, Ngamkhae N, Srimongkolpithak N, Vivithanaporn P, Hongeng S, Thitithanyanont A, Khemawoot P. In vivo evaluation of Andrographis paniculata and Boesenbergia rotunda extract activity against SARS-CoV-2 Delta variant in Golden Syrian hamsters: Potential herbal alternative for COVID-19 treatment. J Tradit Complement Med 2024; 14:598-610. [PMID: 39850600 PMCID: PMC11752117 DOI: 10.1016/j.jtcme.2024.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 01/25/2025] Open
Abstract
The ongoing COVID-19 pandemic has triggered extensive research, mainly focused on identifying effective therapeutic agents, specifically those targeting highly pathogenic SARS-CoV-2 variants. This study aimed to investigate the in vivo antiviral efficacy and anti-inflammatory activity of herbal extracts derived from Andrographis paniculata and Boesenbergia rotunda, using a Golden Syrian hamster model infected with Delta, a representative variant associated with severe COVID-19. Hamsters were intranasally inoculated with the SARS-CoV-2 Delta variant and orally administered either vehicle control, B. rotunda, or A. paniculata extract at a dosage of 1000 mg/kg/day. Euthanasia was conducted on days 1, 3, and 7 post-inoculation, with 4 animals per group. The results demonstrated that oral administration of A. paniculata extract significantly alleviated both lethality and infection severity compared with the vehicle control and B. rotunda extract. However, neither extract exhibited direct antiviral activity in terms of reducing viral load in the lungs. Nonetheless, A. paniculata extract treatment significantly reduced IL-6 protein levels in the lung tissue (7278 ± 868.4 pg/g tissue) compared to the control (12,495 ± 1118 pg/g tissue), indicating there was a decrease in local inflammation. This finding is evidenced by the ability of A. paniculata extract to reduce histological lesions in the lungs of infected hamsters. Furthermore, both extracts significantly decreased IL-6 and IP-10 mRNA expression in peripheral blood mononuclear cells of infected hamsters compared to the control group, suggesting systemic anti-inflammatory effects occurred. In conclusion, A. paniculata extract's potential therapeutic application for SARS-CoV-2 arises from its observed capacity to lessen inflammatory cytokine concentrations and mitigate lung pathology.
Collapse
Affiliation(s)
- Supasek Kongsomros
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| | - Tussapon Boonyarattanasoonthorn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| | - Wallaya Phongphaew
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Chaiyan Kasorndorkbua
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Piyanate Sunyakumthorn
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, 10400, Thailand
| | - Rawiwan Im-Erbsin
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, 10400, Thailand
| | - Luis A. Lugo-Roman
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, 10400, Thailand
| | - Teetat Kongratanapasert
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Jiraporn Paha
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Pakakrong Kwankhao
- Chao Phya Abhaibhubejhr Hospital Foundation, Prachinburi, 25000, Thailand
| | | | - Nittaya Ngamkhae
- Chao Phya Abhaibhubejhr Hospital Foundation, Prachinburi, 25000, Thailand
| | - Nitipol Srimongkolpithak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | | | - Phisit Khemawoot
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn, 10540, Thailand
| |
Collapse
|
32
|
Siddique AI, Sarmah N, Bali NK, Nausch N, Borkakoty B. Differential Gene Expression and Transcriptomics Reveal High M-Gene Expression in JN.1 and KP.1/2 Omicron Sub-Variants of SARS-CoV-2: Implications for Developing More Sensitive Diagnostic Tests. J Med Virol 2024; 96:e70074. [PMID: 39588700 DOI: 10.1002/jmv.70074] [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: 07/31/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024]
Abstract
SARS-CoV-2, a positive-strand RNA virus, utilizes both genomic replication and subgenomic mRNA transcription. Whole genome sequencing (WGS) from clinical samples can estimate viral gene expression levels. WGS was conducted on 529 SARS-CoV-2 positive clinical samples from Assam and northeastern India to track viral emergence and assess gene expression patterns. The results reveal differential expression across structural, non-structural, and accessory genes, with notable upregulation of the M gene, especially in the Omicron variant, followed by E and ORF6. The mean transcript per million (TPM) expression levels of the M gene were significantly higher in Omicron variants (175 611 ± 46 921), peaking in the KP.1/KP.2 sublineage (220 493 ± 34 917), compared to the Delta variant (129 717 ± 33 773). The relative fold change of M gene expression between Delta and Omicron 2024 subvariants showed a 1.6-fold change. Variant-wise gene expression analysis suggests a correlation between gene expression and viral mutation, impacting replication. As anticipated, the expression levels of genes surge with the increase in the virus mutation. The Chi-square trend for average substitution count versus average TPM of the M gene was highly significant (72.78., p < 0.0001). The M gene's high expression and low mutation rate make it an ideal target for designing a real-time RT-PCR kit assay. These findings highlight the need for continuous surveillance and understanding of viral gene expression dynamics for effective COVID-19 management. Further studies are necessary to elucidate the significance of these observations in viral pathogenesis and transmission dynamics.
Collapse
Affiliation(s)
- Aktarul Islam Siddique
- Regional VRDL, Indian Council of Medical Research-Regional Medical Research Centre for NE Region (ICMR-RMRC NE), Dibrugarh, Assam, India
| | - Neelanjana Sarmah
- Regional VRDL, Indian Council of Medical Research-Regional Medical Research Centre for NE Region (ICMR-RMRC NE), Dibrugarh, Assam, India
| | - Nargis K Bali
- Department of Clinical Microbiology, Sher-I Kashmir Institute of Medical Sciences, Soura, Srinagar, Jammu & Kashmir, India
| | - Norman Nausch
- German Epidemic Preparedness Team-SEEG, GIZ, Bonn, Germany
| | - Biswajyoti Borkakoty
- Regional VRDL, Indian Council of Medical Research-Regional Medical Research Centre for NE Region (ICMR-RMRC NE), Dibrugarh, Assam, India
| |
Collapse
|
33
|
Ideguchi S, Miyagi K, Kami W, Tasato D, Higa F, Maeshiro N, Nagamine S, Nakamura H, Kinjo T, Nakamatsu M, Haranaga S, Tokushige A, Ueda S, Fujita J, Yamamoto K. Clinical features of and severity risk factors for COVID-19 in adults during the predominance of SARS-CoV-2 XBB variants in Okinawa, Japan. PLoS One 2024; 19:e0309808. [PMID: 39480756 PMCID: PMC11527311 DOI: 10.1371/journal.pone.0309808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/20/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Since 2023, COVID-19 induced by SARS-CoV-2 XBB variants have been a global epidemic. The XBB variant-induced epidemic was largest in the Okinawa Prefecture among areas in Japan, and healthcare institutions have been burdened by increased COVID-19 hospitalizations. This study aimed to evaluate the clinical features of XBB variant-induced COVID-19 and risk factors for severe COVID-19. METHODS This retrospective study included adult patients hospitalized for COVID-19 between May and July 2023 at four tertiary medical institutions in Okinawa, Japan. Patients with bacterial infection-related complications were excluded. According to oxygen supplementation and intensive care unit admission, patients were divided into two groups, mild and severe. Patient backgrounds, symptoms, and outcomes were compared between both groups, and the risk factors for severe COVID-19 were analyzed using a multivariate logistic regression model. RESULTS In total of 367 patients included, the median age was 75 years, with 18.5% classified into the severe group. The all-cause mortality rate was 4.9%. Patients in the severe group were more older, had more underlying diseases, and had a higher mortality rate (13.2%) than those in the mild group (3.0%). Multivariate logistic regression analysis showed that diabetes mellitus was an independent risk factor for severe COVID-19 (95% confidence interval [CI], 1.002-3.772), whereas bivalent omicron booster vaccination was an independent factor for less severe COVID-19 (95% CI, 0.203-0.862). CONCLUSION This study implies that assessing risk factors in older adults is particularly important in the era of omicron variants.
Collapse
Affiliation(s)
- Shuhei Ideguchi
- Division of Infectious, Respiratory, and Digestive Medicine, First Department of Internal Medicine, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Kazuya Miyagi
- Division of Infectious, Respiratory, and Digestive Medicine, First Department of Internal Medicine, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Wakaki Kami
- Department of Respiratory Medicine, Ohama Dai-ichi Hospital, Okinawa, Japan
| | - Daisuke Tasato
- Department of Respiratory and Infectious Diseases, Okinawa North Medical Association Hospital, Okinawa, Japan
| | - Futoshi Higa
- Department of Respiratory Medicine, NHO Okinawa Hospital, Okinawa, Japan
| | - Noriyuki Maeshiro
- Research Center for Infectious Diseases, Okinawa Prefectural Institute of Health and Environment, Okinawa, Japan
| | - Shota Nagamine
- Research Center for Infectious Diseases, Okinawa Prefectural Institute of Health and Environment, Okinawa, Japan
| | - Hideta Nakamura
- Division of Infectious, Respiratory, and Digestive Medicine, First Department of Internal Medicine, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Takeshi Kinjo
- Division of Infectious, Respiratory, and Digestive Medicine, First Department of Internal Medicine, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Masashi Nakamatsu
- Department of Infection Control, University of the Ryukyus Hospital, Okinawa, Japan
| | - Shusaku Haranaga
- University of the Ryukyus Comprehensive Health Professions Education Center, University Hospital, Okinawa, Japan
| | - Akihiro Tokushige
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Jiro Fujita
- Department of Respiratory Medicine, Ohama Dai-ichi Hospital, Okinawa, Japan
| | - Kazuko Yamamoto
- Division of Infectious, Respiratory, and Digestive Medicine, First Department of Internal Medicine, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| |
Collapse
|
34
|
Chrysostomou AC, Kostrikis LG. Into the Cauldron of the Variant Soup: Insights into the Molecular Epidemiology and Transition to Endemicity of SARS-CoV-2 in Cyprus (November 2022-February 2024). Viruses 2024; 16:1686. [PMID: 39599801 PMCID: PMC11599100 DOI: 10.3390/v16111686] [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: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, driven by the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been characterized by the virus's ongoing evolution, leading to the appearance of more transmissible variants that have often triggered infection surges. In this study, we analyzed the SARS-CoV-2 epidemic in Cyprus, utilizing 1627 viral sequences from infected individuals between November 2022 and February 2024. Over this period, 251 distinct lineages and sublineages were identified, predominantly categorized into three groups: Omicron 5, XBB, and JN.1 (parental lineage BA.2.86), all of which harbor S protein mutations linked to enhanced transmissibility and immune escape. Despite the relatively low numbers of new infections during this period, and the lack of any major waves, unlike earlier phases of the pandemic, these lineages demonstrated varying periods of dominance, with Omicron 5 prevailing from November 2022 to February 2023, XBB variants leading from March to November 2023, and JN.1 generating a wavelet from December 2023 to February 2024. These findings suggest that the SARS-CoV-2 epidemic in Cyprus has reached endemicity, with new variants gradually replacing previously circulating variants irrespective of seasonal patterns. This study highlights the critical importance of ongoing surveillance of SARS-CoV-2 evolution in Cyprus and emphasizes the role of preventive measures in limiting virus transmission, providing valuable insights for safeguarding public health.
Collapse
Affiliation(s)
| | | | - Leondios G. Kostrikis
- Department of Biological Sciences, University of Cyprus, Aglantzia, 2109 Nicosia, Cyprus
- Cyprus Academy of Sciences, Letters, and Arts, 60-68 Phaneromenis Street, 1011 Nicosia, Cyprus
| |
Collapse
|
35
|
Soudani N, Bricker TL, Darling T, Seehra K, Patel N, Guebre-Xabier M, Smith G, Davis-Gardner M, Suthar MS, Ellebedy AH, Boon ACM. Immunogenicity and efficacy of XBB.1.5 rS vaccine against the EG.5.1 variant of SARS-CoV-2 in Syrian hamsters. J Virol 2024; 98:e0052824. [PMID: 39230305 PMCID: PMC11494984 DOI: 10.1128/jvi.00528-24] [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: 03/21/2024] [Accepted: 07/25/2024] [Indexed: 09/05/2024] Open
Abstract
The continued emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants necessitates updating coronavirus disease 2019 (COVID-19) vaccines to match circulating strains. The immunogenicity and efficacy of these vaccines must be tested in pre-clinical animal models. In Syrian hamsters, we measured the humoral and cellular immune response after immunization with the nanoparticle recombinant Spike (S) protein-based COVID-19 vaccine (Novavax, Inc.). We also compared the efficacy of the updated monovalent XBB.1.5 variant vaccine with previous COVID-19 vaccines for the induction of XBB.1.5 and EG.5.1 neutralizing antibodies and protection against a challenge with the EG.5.1 variant of SARS-CoV-2. Immunization induced high levels of S-specific IgG and IgA antibody-secreting cells and antigen-specific CD4+ T cells. The XBB.1.5 and XBB.1.16 vaccines, but not the Prototype vaccine, induced high levels of neutralizing antibodies against the XBB.1.5, EG.5.1, and JN.1 variants of SARS-CoV-2. Upon challenge with the Omicron EG.5.1 variant, the XBB.1.5 and XBB.1.16 vaccines reduced the virus load in the lungs, nasal turbinates, trachea, and nasal washes. The bivalent vaccine (Prototype rS + BA.5 rS) continued to offer protection in the trachea and lungs, but protection was reduced in the upper airways. By contrast, the monovalent Prototype vaccine no longer offered good protection, and breakthrough infections were observed in all animals and tissues. Thus, based on these study results, the protein-based XBB.1.5 vaccine is immunogenic and increased the breadth of protection against the Omicron EG.5.1 variant in the Syrian hamster model. IMPORTANCE As SARS-CoV-2 continues to evolve, there is a need to assess the immunogenicity and efficacy of updated vaccines against newly emerging variants in pre-clinical models such as mice and hamsters. Here, we compared the immunogenicity and efficacy between the updated XBB.1.5, the original Prototype Wuhan-1, and the bivalent Prototype + BA.5 vaccine against a challenge with the EG.5.1 Omicron variant of SARS-CoV-2 in hamsters. The XBB.1.5 and bivalent vaccine, but not the Prototype, induced serum-neutralizing antibodies against EG.5.1, albeit the titers were higher in the XBB.1.5 immunized hamsters. The presence of neutralizing antibodies was associated with complete protection against EG.5.1 infection in the lower airways and reduced virus titers in the upper airways. Compared with the bivalent vaccine, immunization with XBB.1.5 improved viral control in the nasal turbinates. Together, our data show that the updated vaccine is immunogenic and that it offers better protection against recent variants of SARS-CoV-2.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19/virology
- Mesocricetus
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Cricetinae
- Immunogenicity, Vaccine
- Disease Models, Animal
- Vaccine Efficacy
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Female
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
Collapse
Affiliation(s)
- Nadia Soudani
- 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
| | - Traci L. Bricker
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tamarand Darling
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kuljeet Seehra
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nita Patel
- Novavax Inc., Gaithersburg, Maryland, USA
| | | | - Gale Smith
- Novavax Inc., Gaithersburg, Maryland, USA
| | - Meredith Davis-Gardner
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory National Primate Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S. Suthar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory Vaccine Center, Emory National Primate Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Adrianus C. M. Boon
- 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 Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
36
|
Paul D, Verma J, Kumar S, Talukdar D, Jana P, Narendrakumar L, Kumar R, Tanwar S, Gosain M, Porey Karmakar S, Pareek M, Mani S, Chaudhuri S, Kshetrapal P, Wadhwa N, Bhatnagar S, Garg PK, Das B. A rapid point-of-care population-scale dipstick assay to identify and differentiate SARS-CoV-2 variants in COVID-19-positive patients. Front Microbiol 2024; 15:1459644. [PMID: 39498137 PMCID: PMC11532176 DOI: 10.3389/fmicb.2024.1459644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Delta and Omicron variants of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are remarkably contagious, and have been recognized as variants of concern (VOC). The acquisition of spontaneous substitutions or insertion-deletion mutations (indels) in the spike protein-encoding gene substantially increases the binding affinity of the receptor binding domain (RBD)-hACE2 complex and upsurges the transmission of both variants. In this study, we analyzed thousands of genome sequences from 30 distinct SARS-CoV-2 variants, focusing on the unique nucleic acid signatures in the spike gene specific to the Delta and Omicron variants. Using these variant-specific sequences, we synthesized a range of oligonucleotides and optimized a multiplex PCR (mPCR) assay capable of accurately identifying and differentiating between the Delta and Omicron variants. Building on this mPCR assay, we developed a dipstick format by incorporating a tag linker sequence at the 5' end of the forward primer and adding biotin to the 3' end of the oligonucleotides, enhancing the assay's usability and accessibility. Streptavidin-coated latex beads and the dipstick imprinted with a probe for the tag linker sequence in the test strips were used for the detection assay. Our dipstick-based assay, developed as a rapid point-of-care test for identifying and differentiating SARS-CoV-2 variants has the potential to be used in low-resource settings and scaled up to the population level.
Collapse
Affiliation(s)
- Deepjyoti Paul
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Jyoti Verma
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Shakti Kumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Daizee Talukdar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Pradipta Jana
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Lekshmi Narendrakumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Roshan Kumar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Subhash Tanwar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Mudita Gosain
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Sonali Porey Karmakar
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Madhu Pareek
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Shailendra Mani
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Susmita Chaudhuri
- Multidisciplinary Clinical and Translational Research, Translational Health Science and Technology Institute, Faridabad, India
| | - Pallavi Kshetrapal
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Nitya Wadhwa
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Shinjini Bhatnagar
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Pramod Kumar Garg
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Centre for Microbial Research, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
37
|
Yajima H, Nomai T, Okumura K, Maenaka K, Ito J, Hashiguchi T, Sato K. Molecular and structural insights into SARS-CoV-2 evolution: from BA.2 to XBB subvariants. mBio 2024; 15:e0322023. [PMID: 39283095 PMCID: PMC11481514 DOI: 10.1128/mbio.03220-23] [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] [Indexed: 10/19/2024] Open
Abstract
Due to the incessant emergence of various SARS-CoV-2 variants with enhanced fitness in the human population, controlling the COVID-19 pandemic has been challenging. Understanding how the virus enhances its fitness during a pandemic could offer valuable insights for more effective control of viral epidemics. In this manuscript, we review the evolution of SARS-CoV-2 from early 2022 to the end of 2023-from Omicron BA.2 to XBB descendants. Focusing on viral evolution during this period, we provide concrete examples that SARS-CoV-2 has increased its fitness by enhancing several functions of the spike (S) protein, including its binding affinity to the ACE2 receptor and its ability to evade humoral immunity. Furthermore, we explore how specific mutations modify these functions of the S protein through structural alterations. This review provides evolutionary, molecular, and structural insights into how SARS-CoV-2 has increased its fitness and repeatedly caused epidemic surges during the pandemic.
Collapse
Affiliation(s)
- Hisano Yajima
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tomo Nomai
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Kaho Okumura
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Faculty of Liberal Arts, Sophia University, Tokyo, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development, HU-IVReD, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - The Genotype to Phenotype Japan (G2P-Japan) ConsortiumMatsunoKeita1NaoNaganori1SawaHirofumi1MizumaKeita1LiJingshu1KidaIzumi1MimuraYume1OhariYuma1TanakaShinya1TsudaMasumi1WangLei1OdaYoshikata1FerdousZannatul1ShishidoKenji1MohriHiromi1IidaMiki1FukuharaTakasuke1TamuraTomokazu1SuzukiRigel1SuzukiSaori1TsujinoShuhei1ItoHayato1KakuYu2MisawaNaoko2PlianchaisukArnon2GuoZiyi2HinayAlfredo A.Jr.2UsuiKaoru2SaikruangWilaiporn2LytrasSpyridon2UriuKeiya2YoshimuraRyo2KawakuboShusuke2NishumuraLuca2KosugiYusuke2FujitaShigeru2M.TolentinoJarel Elgin2ChenLuo2PanLin2LiWenye2YoMaximilian Stanley2HorinakaKio2SuganamiMai2ChibaMika2YasudaKyoko2IidaKeiko2StrangeAdam Patrick2OhsumiNaomi2TanakaShiho2OgawaEiko2FukudaTsuki2OsujoRina2YoshimuraKazuhisa3SadamasKenji3NagashimaMami3AsakuraHiroyuki3YoshidaIsao3NakagawaSo4TakayamaKazuo5HashimotoRina5DeguchiSayaka5WatanabeYukio5NakataYoshitaka5FutatsusakoHiroki5SakamotoAyaka5YasuharaNaoko5SuzukiTateki5KimuraKanako5SasakiJiei5NakajimaYukari5IrieTakashi6KawabataRyoko6Sasaki-TabataKaori7IkedaTerumasa8NasserHesham8ShimizuRyo8BegumMst Monira8JonathanMichael8MugitaYuka8LeongSharee8TakahashiOtowa8UenoTakamasa8MotozonoChihiro8ToyodaMako8SaitoAkatsuki9KosakaAnon9KawanoMiki9MatsubaraNatsumi9NishiuchiTomoko9ZahradnikJiri10AndrikopoulosProkopios10Padilla-BlancoMiguel10KonarAditi10Hokkaido University, Sapporo, JapanDivision of Systems Virology, Department of Microbiology and Immunolog, The Institute of Medical Science, The University of Tokyo, Tokyo, JapanTokyo Metropolitan Institute of Public Health, Tokyo, JapanTokai University, Kanagawa, JapanKyoto University, Kyoto, JapanHiroshima University, Hiroshima, JapanKyushu University, Fukuoka, JapanKumamoto University, Kumamoto, JapanUniversity of Miyazaki, Miyazaki, JapanCharles University, Vestec-Prague, Czechia
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Faculty of Liberal Arts, Sophia University, Tokyo, Japan
- Institute for Vaccine Research and Development, HU-IVReD, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Kyoto University Immunomonitoring Center, Kyoto University, Kyoto, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Jumpei Ito
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Kyoto University Immunomonitoring Center, Kyoto University, Kyoto, Japan
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| |
Collapse
|
38
|
Arévalo-Romero JA, López-Cantillo G, Moreno-Jiménez S, Marcos-Alcalde Í, Ros-Pardo D, Camacho BA, Gómez-Puertas P, Ramírez-Segura CA. In Silico Design of miniACE2 Decoys with In Vitro Enhanced Neutralization Activity against SARS-CoV-2, Encompassing Omicron Subvariants. Int J Mol Sci 2024; 25:10802. [PMID: 39409131 PMCID: PMC11476394 DOI: 10.3390/ijms251910802] [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: 09/14/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
The COVID-19 pandemic has overwhelmed healthcare systems and triggered global economic downturns. While vaccines have reduced the lethality rate of SARS-CoV-2 to 0.9% as of October 2024, the continuous evolution of variants remains a significant public health challenge. Next-generation medical therapies offer hope in addressing this threat, especially for immunocompromised individuals who experience prolonged infections and severe illnesses, contributing to viral evolution. These cases increase the risk of new variants emerging. This study explores miniACE2 decoys as a novel strategy to counteract SARS-CoV-2 variants. Using in silico design and molecular dynamics, blocking proteins (BPs) were developed with stronger binding affinity for the receptor-binding domain of multiple variants than naturally soluble human ACE2. The BPs were expressed in E. coli and tested in vitro, showing promising neutralizing effects. Notably, miniACE2 BP9 exhibited an average IC50 of 4.9 µg/mL across several variants, including the Wuhan strain, Mu, Omicron BA.1, and BA.2 This low IC50 demonstrates the potent neutralizing ability of BP9, indicating its efficacy at low concentrations.Based on these findings, BP9 has emerged as a promising therapeutic candidate for combating SARS-CoV-2 and its evolving variants, thereby positioning it as a potential emergency biopharmaceutical.
Collapse
Affiliation(s)
- Jenny Andrea Arévalo-Romero
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Gina López-Cantillo
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| | - Sara Moreno-Jiménez
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| | - Íñigo Marcos-Alcalde
- Grupo de Modelado Molecular del Centro de Biología Molecular Severo Ochoa, 14 CSIC-UAM, 28049 Madrid, Spain; (Í.M.-A.); (D.R.-P.)
| | - David Ros-Pardo
- Grupo de Modelado Molecular del Centro de Biología Molecular Severo Ochoa, 14 CSIC-UAM, 28049 Madrid, Spain; (Í.M.-A.); (D.R.-P.)
| | - Bernardo Armando Camacho
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| | - Paulino Gómez-Puertas
- Grupo de Modelado Molecular del Centro de Biología Molecular Severo Ochoa, 14 CSIC-UAM, 28049 Madrid, Spain; (Í.M.-A.); (D.R.-P.)
| | - Cesar A. Ramírez-Segura
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| |
Collapse
|
39
|
Yajima H, Anraku Y, Kaku Y, Kimura KT, Plianchaisuk A, Okumura K, Nakada-Nakura Y, Atarashi Y, Hemmi T, Kuroda D, Takahashi Y, Kita S, Sasaki J, Sumita H, Ito J, Maenaka K, Sato K, Hashiguchi T. Structural basis for receptor-binding domain mobility of the spike in SARS-CoV-2 BA.2.86 and JN.1. Nat Commun 2024; 15:8574. [PMID: 39375326 PMCID: PMC11458767 DOI: 10.1038/s41467-024-52808-2] [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: 03/15/2024] [Accepted: 09/18/2024] [Indexed: 10/09/2024] Open
Abstract
Since 2019, SARS-CoV-2 has undergone mutations, resulting in pandemic and epidemic waves. The SARS-CoV-2 spike protein, crucial for cellular entry, binds to the ACE2 receptor exclusively when its receptor-binding domain (RBD) adopts the up-conformation. However, whether ACE2 also interacts with the RBD in the down-conformation to facilitate the conformational shift to RBD-up remains unclear. Herein, we present the structures of the BA.2.86 and the JN.1 spike proteins bound to ACE2. Notably, we successfully observed the ACE2-bound down-RBD, indicating an intermediate structure before the RBD-up conformation. The wider and mobile angle of RBDs in the up-state provides space for ACE2 to interact with the down-RBD, facilitating the transition to the RBD-up state. The K356T, but not N354-linked glycan, contributes to both of infectivity and neutralizing-antibody evasion in BA.2.86. These structural insights the spike-protein dynamics would help understand the mechanisms underlying SARS-CoV-2 infection and its neutralization.
Collapse
Affiliation(s)
- Hisano Yajima
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yuki Anraku
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yu Kaku
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kanako Terakado Kimura
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Arnon Plianchaisuk
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kaho Okumura
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Faculty of Liberal Arts, Sophia University, Tokyo, Japan
| | - Yoshiko Nakada-Nakura
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yusuke Atarashi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases; Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Takuya Hemmi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Daisuke Kuroda
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases; Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases; Shinjuku-ku, Tokyo, 162-8640, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Shunsuke Kita
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Jiei Sasaki
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiromi Sumita
- Research Administration Office, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jumpei Ito
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
- Kyushu University, Fukuoka, Japan
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan.
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan.
- Kyoto University Immunomonitoring Center, Kyoto University, Kyoto, Japan.
| |
Collapse
|
40
|
Djorwé S, Malki A, Nzoyikorera N, Nyandwi J, Zebsoubo SP, Bellamine K, Bousfiha A. Genetic diversity and genomic epidemiology of SARS-CoV-2 during the first 3 years of the pandemic in Morocco: comprehensive sequence analysis, including the unique lineage B.1.528 in Morocco. Access Microbiol 2024; 6:000853.v4. [PMID: 39376591 PMCID: PMC11457919 DOI: 10.1099/acmi.0.000853.v4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
During the 3 years following the emergence of the COVID-19 pandemic, the African continent, like other regions of the world, was substantially impacted by COVID-19. In Morocco, the COVID-19 pandemic has been marked by the emergence and spread of several SARS-CoV-2 variants, leading to a substantial increase in the incidence of infections and deaths. Nevertheless, the comprehensive understanding of the genetic diversity, evolution, and epidemiology of several viral lineages remained limited in Morocco. This study sought to deepen the understanding of the genomic epidemiology of SARS-CoV-2 through a retrospective analysis. The main objective of this study was to analyse the genetic diversity of SARS-CoV-2 and identify distinct lineages, as well as assess their evolution during the pandemic in Morocco, using genomic epidemiology approaches. Furthermore, several key mutations in the functional proteins across different viral lineages were highlighted along with an analysis of the genetic relationships amongst these strains to better understand their evolutionary pathways. A total of 2274 genomic sequences of SARS-CoV-2 isolated in Morocco during the period of 2020 to 2023, were extracted from the GISAID EpiCoV database and subjected to analysis. Lineages and clades were classified according to the nomenclature of GISAID, Nextstrain, and Pangolin. The study was conducted and reported in accordance with STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) guidelines. An exhaustive analysis of 2274 genomic sequences led to the identification of 157 PANGO lineages, including notable lineages such as B.1, B.1.1, B.1.528, and B.1.177, as well as variants such as B.1.1.7, B.1.621, B.1.525, B.1.351, B.1.617.1, B.1.617.2, and its notable sublineages AY.33, AY.72, AY.112, AY.121 that evolved over time before being supplanted by Omicron in December 2021. Among the 2274 sequences analysed, Omicron and its subvariants had a prevalence of 59.5%. The most predominant clades were 21K, 21L, and 22B, which are respectively related phylogenetically to BA.1, BA.2, and BA.5. In June 2022, Morocco rapidly observed a recrudescence of cases of infection, with the emergence and concurrent coexistence of subvariants from clade 22B such as BA.5.2.20, BA.5, BA.5.1, BA.5.2.1, and BF.5, supplanting the subvariants BA.1 (clade display 21K) and BA.2 (clade display 21L), which became marginal. However, XBB (clade 22F) and its progeny such XBB.1.5(23A), XBB.1.16(23B), CH.1.1(23C), XBB.1.9(23D), XBB.2.3(23E), EG.5.1(23F), and XBB.1.5.70(23G) have evolved sporadically. Furthermore, several notable mutations, such as H69del/V70del, G142D, K417N, T478K, E484K, E484A, L452R, F486P, N501Y, Q613H, D614G, and P681H/R, have been identified. Some of these SARS-CoV-2 mutations are known to be involved in increasing transmissibility, virulence, and antibody escape. This study has identified several distinct lineages and mutations involved in the genetic diversity of Moroccan isolates, as well as the analysis of their evolutionary trends. These findings provide a robust basis for better understanding the distinct mutations and their roles in the variation of transmissibility, pathogenicity, and antigenicity (immune evasion/reinfection). Furthermore, the noteworthy number of distinct lineages identified in Morocco highlights the importance of maintaining continuous surveillance of COVID-19. Moreover, expanding vaccination coverage would also help protect patients against more severe clinical disease.
Collapse
Affiliation(s)
- Soulandi Djorwé
- Laboratory of Physiopathology and Molecular Genetics, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca (Morocco), Avenue Cdt Driss El Harti, PB 7955 Sidi Othman, Casablanca, Morocco
- Bourgogne Laboratory of Medical and Scientific Analysis, 136, residence belhcen, Bd Bourgogne, Casablanca, Morocco
| | - Abderrahim Malki
- Laboratory of Physiopathology and Molecular Genetics, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca (Morocco), Avenue Cdt Driss El Harti, PB 7955 Sidi Othman, Casablanca, Morocco
| | - Néhémie Nzoyikorera
- National Reference Laboratory, National Institute of Public Health, Bujumbura, Burundi
- Higher Institute of Biosciences and Biotechnology, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
- Laboratory of Microbial Biotechnology and Infectiology Research, Mohammed VI Center for Research & Innovation, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco
| | - Joseph Nyandwi
- Département de Médecine, Faculté de Médecine, Université du Burundi, Bujumbura, Burundi
- Ministère de la Santé Publique et de la Lutte contre le Sida, Institut National de Santé Publique de Bujumbura, Bujumbura, Burundi
| | - Samuel Privat Zebsoubo
- School of Advanced Studies in Biotechnology and Private Health (EHEB), 183 Bd de la Resistance, Casablanca 20250, Morocco
| | - Kawthar Bellamine
- Bourgogne Laboratory of Medical and Scientific Analysis, 136, residence belhcen, Bd Bourgogne, Casablanca, Morocco
| | - Amale Bousfiha
- Laboratory of Physiopathology and Molecular Genetics, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca (Morocco), Avenue Cdt Driss El Harti, PB 7955 Sidi Othman, Casablanca, Morocco
| |
Collapse
|
41
|
Zhang L, Tian S, Dai J, Li Y, Zhou Y, Li Y, Xu J, Liu S, Lin Z, Zhang Z, Chen J, Wei P, Zhao J, Jin J, Wang Y, Zhao J. Trivalent SARS-CoV-2 virus-like particle vaccines exhibit broad-spectrum neutralization and protection against XBB.1 and BA.2.86 variants. Virol Sin 2024; 39:836-839. [PMID: 39173911 PMCID: PMC11738757 DOI: 10.1016/j.virs.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
•RBDs of the WT, BQ.1.1 and XBB.1 variants were genetically fused and displayed on the VLP to create RBDV14 M. •RBDV14 M can induce superior antibody responses against the newly emerged SARS-CoV-2 variants XBB.1, EG.5 and BA.2.86. •RBDV14 M is one of the few existing next-generation vaccine candidates that utilize virus-like particle platform.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory of Respiratory Disease, Public Health Safety Center Laboratory of General Administration of Customs, Guangzhou Customs Technology Center, Guangzhou, Guangdong, 510182, China
| | - Siyu Tian
- Patronus Biotech Co. Ltd., Guangzhou, 510182, China
| | - Jun Dai
- State Key Laboratory of Respiratory Disease, Public Health Safety Center Laboratory of General Administration of Customs, Guangzhou Customs Technology Center, Guangzhou, Guangdong, 510182, China; 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, Guangdong, 510182, China; Guangzhou National Laboratory, Guangzhou, Guangdong, 510182, China
| | - Yuanyuan Li
- Patronus Biotech Co. Ltd., Guangzhou, 510182, China
| | - Yu Zhou
- Patronus Biotech Co. Ltd., Guangzhou, 510182, China
| | - Yan Li
- Patronus Biotech Co. Ltd., Guangzhou, 510182, China
| | - Jiao Xu
- Patronus Biotech Co. Ltd., Guangzhou, 510182, China
| | - Shuyun Liu
- Patronus Biotech Co. Ltd., Guangzhou, 510182, China
| | - Zhiwei Lin
- State Key Laboratory of Respiratory Disease, Public Health Safety Center Laboratory of General Administration of Customs, Guangzhou Customs Technology Center, Guangzhou, Guangdong, 510182, China
| | - Zhaoyong Zhang
- 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, Guangdong, 510182, China
| | - Jiantao Chen
- 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, Guangdong, 510182, China
| | - Peilan Wei
- 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, Guangdong, 510182, China; Guangzhou National Laboratory, Guangzhou, Guangdong, 510182, China
| | - Jingxian Zhao
- 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, Guangdong, 510182, China; Guangzhou National Laboratory, Guangzhou, Guangdong, 510182, China.
| | - Jing Jin
- Patronus Biotech Co. Ltd., Guangzhou, 510182, 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, Guangdong, 510182, China; Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| | - Jincun Zhao
- 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, Guangdong, 510182, China; Guangzhou National Laboratory, Guangzhou, Guangdong, 510182, China; Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China.
| |
Collapse
|
42
|
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
|
43
|
Ruiz F, Foreman WB, Lilly M, Baharani VA, Depierreux DM, Chohan V, Taylor AL, Guenthoer J, Ralph D, Matsen IV FA, Chu HY, Bieniasz PD, Côté M, Starr TN, Overbaugh J. Delineating the functional activity of antibodies with cross-reactivity to SARS-CoV-2, SARS-CoV-1 and related sarbecoviruses. PLoS Pathog 2024; 20:e1012650. [PMID: 39466880 PMCID: PMC11542851 DOI: 10.1371/journal.ppat.1012650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/07/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
The recurring spillover of pathogenic coronaviruses and demonstrated capacity of sarbecoviruses, such SARS-CoV-2, to rapidly evolve in humans underscores the need to better understand immune responses to this virus family. For this purpose, we characterized the functional breadth and potency of antibodies targeting the receptor binding domain (RBD) of the spike glycoprotein that exhibited cross-reactivity against SARS-CoV-2 variants, SARS-CoV-1 and sarbecoviruses from diverse clades and animal origins with spillover potential. One neutralizing antibody, C68.61, showed remarkable neutralization breadth against both SARS-CoV-2 variants and viruses from different sarbecovirus clades. C68.61, which targets a conserved RBD class 5 epitope, did not select for escape variants of SARS-CoV-2 or SARS-CoV-1 in culture nor have predicted escape variants among circulating SARS-CoV-2 strains, suggesting this epitope is functionally constrained. We identified 11 additional SARS-CoV-2/SARS-CoV-1 cross-reactive antibodies that target the more sequence conserved class 4 and class 5 epitopes within RBD that show activity against a subset of diverse sarbecoviruses with one antibody binding every single sarbecovirus RBD tested. A subset of these antibodies exhibited Fc-mediated effector functions as potent as antibodies that impact infection outcome in animal models. Thus, our study identified antibodies targeting conserved regions across SARS-CoV-2 variants and sarbecoviruses that may serve as therapeutics for pandemic preparedness as well as blueprints for the design of immunogens capable of eliciting cross-neutralizing responses.
Collapse
Affiliation(s)
- Felicitas Ruiz
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, United States of America
| | - William B. Foreman
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Michelle Lilly
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Viren A. Baharani
- Laboratory of Retrovirology, The Rockefeller University, New York, New York, United States of America
- Laboratory of Molecular Immunology, The Rockefeller University, New York, New York, United States of America
| | - Delphine M. Depierreux
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Vrasha Chohan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Ashley L. Taylor
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Jamie Guenthoer
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Duncan Ralph
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Frederick A. Matsen IV
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
| | - Helen Y. Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, United States of America
| | - Paul D. Bieniasz
- Laboratory of Retrovirology, The Rockefeller University, New York, New York, United States of America
- Howard Hughes Medical Institute, The Rockefeller University, New York, New York, United States of America
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, and Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
| | - Tyler N. Starr
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| |
Collapse
|
44
|
Kustova DD, Pochtovyi AA, Shpakova OG, Shtinova IA, Kuznetsova NA, Kleimenov DA, Komarov AG, Gushchin VA. [The Molecular and Biological Patterns Underlying Sustained SARS-CoV-2 Circulation in the Human Population]. Vopr Virusol 2024; 69:329-340. [PMID: 39361927 DOI: 10.36233/0507-4088-242] [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: 06/16/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION For four years, SARS-CoV-2, the etiological agent of COVID-19, has been circulating among humans. By the end of the second year, an absence of immunologically naive individuals was observed, attributable to extensive immunization efforts and natural viral exposure. This study focuses on delineating the molecular and biological patterns that facilitate the persistence of SARS-CoV-2, thereby informing predictions on the epidemiological trajectory of COVID-19 toward refining pandemic countermeasures. The aim of this study was to describe the molecular biological patterns identified that contribute to the persistence of the virus in the human population. MATERIALS AND METHODS For over three years since the beginning of the COVID-19 pandemic, molecular genetic monitoring of SARS-CoV-2 has been conducted, which included the collection of nasopharyngeal swabs from infected individuals, assessment of viral load, and subsequent whole-genome sequencing. RESULTS We discerned dominant genetic lineages correlated with rising disease incidence. We scrutinized amino acid substitutions across SARS-CoV-2 proteins and quantified viral loads in swab samples from patients with emerging COVID-19 variants. Our findings suggest a model of viral persistence characterized by 1) periodic serotype shifts causing substantial diminutions in serum virus-neutralizing activity (> 10-fold), 2) serotype-specific accrual of point mutations in the receptor-binding domain (RBD) to modestly circumvent neutralizing antibodies and enhance receptor affinity, and 3) a gradually increasing amount of virus being shed in mucosal surfaces within a single serotype. CONCLUSION This model aptly accounts for the dynamics of COVID-19 incidence in Moscow. For a comprehensive understanding of these dynamics, acquiring population-level data on immune tension and antibody neutralization relative to genetic lineage compositions is essential.
Collapse
Affiliation(s)
- D D Kustova
- National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
- Federal State Budgetary Educational Institution of Higher Education Lomonosov Moscow State University
| | - A A Pochtovyi
- National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
- Federal State Budgetary Educational Institution of Higher Education Lomonosov Moscow State University
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
| | | | | | - N A Kuznetsova
- National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
| | - D A Kleimenov
- National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
| | | | - V A Gushchin
- National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya of the Ministry of Health of the Russian Federation
- Federal State Budgetary Educational Institution of Higher Education Lomonosov Moscow State University
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University)
| |
Collapse
|
45
|
Chen J, Huang Z, Xiao J, Du S, Bu Q, Guo H, Ye J, Chen S, Gao J, Li Z, Lan M, Wang S, Zhang T, Zhang J, Wu Y, Zhang Y, Xia N, Yuan Q, Cheng T. A quadri-fluorescence SARS-CoV-2 pseudovirus system for efficient antigenic characterization of multiple circulating variants. CELL REPORTS METHODS 2024; 4:100856. [PMID: 39243752 PMCID: PMC11440059 DOI: 10.1016/j.crmeth.2024.100856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/18/2024] [Accepted: 08/14/2024] [Indexed: 09/09/2024]
Abstract
The ongoing co-circulation of multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strains necessitates advanced methods such as high-throughput multiplex pseudovirus systems for evaluating immune responses to different variants, crucial for developing updated vaccines and neutralizing antibodies (nAbs). We have developed a quadri-fluorescence (qFluo) pseudovirus platform by four fluorescent reporters with different spectra, allowing simultaneous measurement of the nAbs against four variants in a single test. qFluo shows high concordance with the classical single-reporter assay when testing monoclonal antibodies and human plasma. Utilizing qFluo, we assessed the immunogenicities of the spike of BA.5, BQ.1.1, XBB.1.5, and CH.1.1 in hamsters. An analysis of cross-neutralization against 51 variants demonstrated superior protective immunity from XBB.1.5, especially against prevalent strains such as "FLip" and JN.1, compared to BA.5. Our finding partially fills the knowledge gap concerning the immunogenic efficacy of the XBB.1.5 vaccine against current dominant variants, being instrumental in vaccine-strain decisions and insight into the evolutionary path of SARS-CoV-2.
Collapse
Affiliation(s)
- Jijing Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Zehong Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Jin Xiao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Shuangling Du
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Qingfang Bu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Huilin Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Jianghui Ye
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Shiqi Chen
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jiahua Gao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Zonglin Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Miaolin Lan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Shaojuan Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Tianying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Jiming Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, Shanghai Institute of Infectious Diseases and Biosecurity, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yangtao Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China.
| | - Yali Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China.
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, School of Life Sciences, Xiamen University, Xiamen 361102, P.R. China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen 361102, P.R. China.
| |
Collapse
|
46
|
Holmes EC. The Emergence and Evolution of SARS-CoV-2. Annu Rev Virol 2024; 11:21-42. [PMID: 38631919 DOI: 10.1146/annurev-virology-093022-013037] [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: 04/19/2024]
Abstract
The origin of SARS-CoV-2 has evoked heated debate and strong accusations, yet seemingly little resolution. I review the scientific evidence on the origin of SARS-CoV-2 and its subsequent spread through the human population. The available data clearly point to a natural zoonotic emergence within, or closely linked to, the Huanan Seafood Wholesale Market in Wuhan. There is no direct evidence linking the emergence of SARS-CoV-2 to laboratory work conducted at the Wuhan Institute of Virology. The subsequent global spread of SARS-CoV-2 was characterized by a gradual adaptation to humans, with dual increases in transmissibility and virulence until the emergence of the Omicron variant. Of note has been the frequent transmission of SARS-CoV-2 from humans to other animals, marking it as a strongly host generalist virus. Unless lessons from the origin of SARS-CoV-2 are learned, it is inevitable that more zoonotic events leading to more epidemics and pandemics will plague human populations.
Collapse
Affiliation(s)
- Edward C Holmes
- Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia;
| |
Collapse
|
47
|
Nyame P, Togami A, Yoshida T, Masunaga T, Begum MM, Terasawa H, Monde N, Tahara Y, Tanaka R, Tanaka Y, Appiah-Kubi J, Amesimeku WAO, Hossain MJ, Otsuka M, Yoshimura K, Ikeda T, Sawa T, Satou Y, Fujita M, Maeda Y, Tateishi H, Monde K. A heterocyclic compound inhibits viral release by inducing cell surface BST2/Tetherin/CD317/HM1.24. J Biol Chem 2024; 300:107701. [PMID: 39173946 PMCID: PMC11419809 DOI: 10.1016/j.jbc.2024.107701] [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: 05/31/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024] Open
Abstract
The introduction of combined antiretroviral therapy (cART) has greatly improved the quality of life of human immunodeficiency virus type 1 (HIV-1)-infected individuals. Nonetheless, the ever-present desire to seek out a full remedy for HIV-1 infections makes the discovery of novel antiviral medication compelling. Owing to this, a new late-stage inhibitor, Lenacapavir/Sunlenca, an HIV multi-phase suppressor, was clinically authorized in 2022. Besides unveiling cutting-edge antivirals inhibiting late-stage proteins or processes, newer therapeutics targeting host restriction factors hold promise for the curative care of HIV-1 infections. Notwithstanding, bone marrow stromal antigen 2 (BST2)/Tetherin/CD317/HM1.24, which entraps progeny virions is an appealing HIV-1 therapeutic candidate. In this study, a novel drug screening system was established, using the Jurkat/Vpr-HiBiT T cells, to identify drugs that could obstruct HIV-1 release; the candidate compounds were selected from the Ono Pharmaceutical compound library. Jurkat T cells expressing Vpr-HiBiT were infected with NL4-3, and the amount of virus release was quantified indirectly by the amount of Vpr-HiBiT incorporated into the progeny virions. Subsequently, the candidate compounds that suppressed viral release were used to synthesize the heterocyclic compound, HT-7, which reduces HIV-1 release with less cellular toxicity. Notably, HT-7 increased cell surface BST2 coupled with HIV-1 release reduction in Jurkat cells but not Jurkat/KO-BST2 cells. Seemingly, HT-7 impeded simian immunodeficiency virus (SIV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) release. Concisely, these results suggest that the reduction in viral release, following HT-7 treatment, resulted from the modulation of cell surface expression of BST2 by HT-7.
Collapse
Affiliation(s)
- Perpetual Nyame
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Akihiro Togami
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomofumi Yoshida
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takuya Masunaga
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mst Monira Begum
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Hiromi Terasawa
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Nami Monde
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yurika Tahara
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Reiko Tanaka
- Laboratory of Hemato-Immunology, Graduate School of Health Sciences, University of the Ryukyus, Okinawa, Japan
| | - Yuetsu Tanaka
- Laboratory of Hemato-Immunology, Graduate School of Health Sciences, University of the Ryukyus, Okinawa, Japan
| | - Joyce Appiah-Kubi
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Md Jakir Hossain
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masami Otsuka
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Department of Drug Discovery, Science Farm Ltd, Kumamoto, Japan
| | - Kazuhisa Yoshimura
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yorifumi Satou
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Maeda
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Department of Nursing, Kibi International University, Takahashi, Japan
| | - Hiroshi Tateishi
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Research & Development, Hirata Corporation, Kumamoto, Japan.
| | - Kazuaki Monde
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
48
|
Tsujino S, Deguchi S, Nomai T, Padilla-Blanco M, Plianchaisuk A, Wang L, Begum MM, Uriu K, Mizuma K, Nao N, Kojima I, Tsubo T, Li J, Matsumura Y, Nagao M, Oda Y, Tsuda M, Anraku Y, Kita S, Yajima H, Sasaki-Tabata K, Guo Z, Hinay AA, Yoshimatsu K, Yamamoto Y, Nagamoto T, Asakura H, Nagashima M, Sadamasu K, Yoshimura K, Nasser H, Jonathan M, Putri O, Kim Y, Chen L, Suzuki R, Tamura T, Maenaka K, Irie T, Matsuno K, Tanaka S, Ito J, Ikeda T, Takayama K, Zahradnik J, Hashiguchi T, Fukuhara T, Sato K. Virological characteristics of the SARS-CoV-2 Omicron EG.5.1 variant. Microbiol Immunol 2024; 68:305-330. [PMID: 38961765 DOI: 10.1111/1348-0421.13165] [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: 03/27/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024]
Abstract
In middle to late 2023, a sublineage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron XBB, EG.5.1 (a progeny of XBB.1.9.2), is spreading rapidly around the world. We performed multiscale investigations, including phylogenetic analysis, epidemic dynamics modeling, infection experiments using pseudoviruses, clinical isolates, and recombinant viruses in cell cultures and experimental animals, and the use of human sera and antiviral compounds, to reveal the virological features of the newly emerging EG.5.1 variant. Our phylogenetic analysis and epidemic dynamics modeling suggested that two hallmark substitutions of EG.5.1, S:F456L and ORF9b:I5T are critical to its increased viral fitness. Experimental investigations on the growth kinetics, sensitivity to clinically available antivirals, fusogenicity, and pathogenicity of EG.5.1 suggested that the virological features of EG.5.1 are comparable to those of XBB.1.5. However, cryo-electron microscopy revealed structural differences between the spike proteins of EG.5.1 and XBB.1.5. We further assessed the impact of ORF9b:I5T on viral features, but it was almost negligible in our experimental setup. Our multiscale investigations provide knowledge for understanding the evolutionary traits of newly emerging pathogenic viruses, including EG.5.1, in the human population.
Collapse
Affiliation(s)
- Shuhei Tsujino
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Tomo Nomai
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Miguel Padilla-Blanco
- First Medical Faculty at Biocev, Charles University, Vestec-Prague, Czechia
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU (UCH-CEU), CEU Universities, Valencia, Spain
| | - Arnon Plianchaisuk
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Mst Monira Begum
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Keiya Uriu
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keita Mizuma
- Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Naganori Nao
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Isshu Kojima
- Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Tomoya Tsubo
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Jingshu Li
- Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasufumi Matsumura
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Yuki Anraku
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Shunsuke Kita
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hisano Yajima
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Kaori Sasaki-Tabata
- Department of Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Ziyi Guo
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Alfredo A Hinay
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | - Mami Nagashima
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | - Kenji Sadamasu
- Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | | | - Hesham Nasser
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Michael Jonathan
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Olivia Putri
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Biomedicine, School of Life Sciences, Indonesia International Institute for Life Sciences (i3L), Jakarta, Indonesia
| | - Yoonjin Kim
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Life Sciences, Faculty of Natural Science, Imperial College London, London, UK
| | - Luo Chen
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Tomokazu Tamura
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Irie
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Keita Matsuno
- Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Jumpei Ito
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Jiri Zahradnik
- First Medical Faculty at Biocev, Charles University, Vestec-Prague, Czechia
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
49
|
Yamamoto S, Matsuda K, Maeda K, Mizoue T, Horii K, Okudera K, Tan T, Oshiro Y, Inamura N, Nemoto T, S Takeuchi J, Konishi M, Sugiyama H, Aoyanagi N, Sugiura W, Ohmagari N. Protection of Omicron Bivalent Vaccine, Previous Infection, and Their Induced Neutralizing Antibodies Against Symptomatic Infection With Omicron XBB.1.16 and EG.5.1. Open Forum Infect Dis 2024; 11:ofae519. [PMID: 39319092 PMCID: PMC11420683 DOI: 10.1093/ofid/ofae519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
Background Data are limited on the protective role of the Omicron BA bivalent vaccine, previous infection, and their induced neutralizing antibodies against Omicron XBB.1.16 and EG.5.1 infection. Methods We conducted a nested case-control analysis among tertiary hospital staff in Tokyo who had received ≥3 doses of COVID-19 vaccines and donated blood samples in June 2023 (1 month before the Omicron XBB.1.16 and EG.5.1 wave). We identified 206 symptomatic cases between June and September 2023 and selected their controls with 1:1 propensity score matching. We examined the association of vaccination, previous infection, and preinfection live virus neutralizing antibody titers against Omicron XBB.1.16 and EG.5.1 with the risk of COVID-19 infection. Results Previous infection during the Omicron BA- or XBB-dominant phase was associated with a significantly lower infection risk during the XBB.1.16 and EG.5.1-dominant phase than infection-naive status, with 70% and 100% protection, respectively, whereas Omicron BA bivalent vaccination showed no association. Preinfection neutralizing titers against XBB.1.16 and EG.5.1 were 39% (95% CI, 8%-60%) and 28% (95% CI, 8%-44%) lower in cases than matched controls. Neutralizing activity against XBB.1.16 and EG.5.1 was somewhat detectable in the sera of individuals with previous infection but barely detectable in those who were infection naive and received the Omicron bivalent vaccine. Conclusions In the era when the Omicron XBB vaccine was unavailable, the Omicron BA bivalent vaccine did not confer the neutralizing activity and protection against Omicron XBB.1.16 and EG.5.1 symptomatic infection. The previous infection afforded neutralizing titers and protection against symptomatic infection with these variants.
Collapse
Affiliation(s)
- Shohei Yamamoto
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kouki Matsuda
- Division of Antiviral Therapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
| | - Kenji Maeda
- Division of Antiviral Therapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan
- Department of Refractory Viral Infection, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tetsuya Mizoue
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kumi Horii
- Infection Control Office, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Kaori Okudera
- Infection Control Office, Kohnodai Hospital of the National Center for the Global Health and Medicine, Chiba, Japan
| | - Tomofumi Tan
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Yusuke Oshiro
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Natsumi Inamura
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Takashi Nemoto
- Department of Laboratory Testing, Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Junko S Takeuchi
- Department of Academic-Industrial Partnerships Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Maki Konishi
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Haruhito Sugiyama
- Center Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Nobuyoshi Aoyanagi
- Kohnodai Hospital of the National Center for the Global Health and Medicine, Chiba, Japan
| | - Wataru Sugiura
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Liang Z, Li C, Gong X, Ye G, Jiang Y, Shi H, Hussain A, Zhao M, Li M, Tian Y, Zhao W, Yang Y, Huang Y, Shen C, Yang M. Development of Glycan-masked SARS-CoV-2 RBD vaccines against SARS-related coronaviruses. PLoS Pathog 2024; 20:e1012599. [PMID: 39325829 PMCID: PMC11460674 DOI: 10.1371/journal.ppat.1012599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 10/08/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Emerging and recurrent infectious diseases caused by coronaviruses remain a significant public health concern. Here, we present a targeted approach to elicit antibodies capable of neutralizing SARS-CoV-2 variants and other SARS-related coronaviruses. By introducing amino acid mutations at mutation-prone sites, we engineered glycosylation modifications to the Receptor Binding Domain (RBD) of SARS-CoV-2, thereby exposing more conserved, yet less accessible epitopes. We developed both messenger RNA (mRNA) and recombination subunit vaccines using these engineered-RBDs (M1, M2) and the wild-type RBD as immunogens. The engineered-RBD vaccines elicited robust neutralizing responses against various SARS-CoV-2 variants as well as SARS-CoV and WIV1-CoV, and conferred protection in mice challenged with the XBB.1.16 strain. Furthermore, We highlighted that glycan masking is a decisive factor in antibody binding changes and RBD-conserved antibody response. Additionally, the glycan-engineered RBD mRNA vaccines stimulated stronger cell-mediated immune responses. Our glycan modification strategy significantly enhances broad-spectrum neutralizing efficacy and cellular immunity, providing valuable insights for the development of vaccines against a wide range of SARS-related coronaviruses.
Collapse
Affiliation(s)
- Zuxin Liang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Chunhui Li
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Xiaohua Gong
- National Clinical Research Center for infectious disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People’s Republic of China
- Laboratory Animal Center, Anhui Medical University, Hefei, People’s Republic of China
| | - Guoguo Ye
- National Clinical Research Center for infectious disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Yushan Jiang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Huiping Shi
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Abid Hussain
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Mengyuan Zhao
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Mengjun Li
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Yuxin Tian
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
| | - Yang Yang
- National Clinical Research Center for infectious disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Yuanyu Huang
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People’s Republic of China
| | - Chenguang Shen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health; Department of Laboratory Medicine, Zhujiang Hospital; Southern Medical University, Guangzhou, People’s Republic of China
- Key Laboratory of Infectious Diseases Research in South China, Southern Medical University, Ministry of Education, Guangzhou, People’s Republic of China
| | - Minghui Yang
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, People’s Republic of China
| |
Collapse
|