1
|
Trauer JM, Hughes AE, Shipman DS, Meehan MT, Henderson AS, McBryde ES, Ragonnet R. A data science pipeline applied to Australia's 2022 COVID-19 Omicron waves. Infect Dis Model 2025; 10:99-109. [PMID: 39364337 PMCID: PMC11447346 DOI: 10.1016/j.idm.2024.08.005] [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: 02/29/2024] [Revised: 08/04/2024] [Accepted: 08/22/2024] [Indexed: 10/05/2024] Open
Abstract
The field of software engineering is advancing at astonishing speed, with packages now available to support many stages of data science pipelines. These packages can support infectious disease modelling to be more robust, efficient and transparent, which has been particularly important during the COVID-19 pandemic. We developed a package for the construction of infectious disease models, integrated it with several open-source libraries and applied this composite pipeline to multiple data sources that provided insights into Australia's 2022 COVID-19 epidemic. We aimed to identify the key processes relevant to COVID-19 transmission dynamics and thereby develop a model that could quantify relevant epidemiological parameters. The pipeline's advantages include markedly increased speed, an expressive application programming interface, the transparency of open-source development, easy access to a broad range of calibration and optimisation tools and consideration of the full workflow from input manipulation through to algorithmic generation of the publication materials. Extending the base model to include mobility effects slightly improved model fit to data, with this approach selected as the model configuration for further epidemiological inference. Under our assumption of widespread immunity against severe outcomes from recent vaccination, incorporating an additional effect of the main vaccination programs rolled out during 2022 on transmission did not further improve model fit. Our simulations suggested that one in every two to six COVID-19 episodes were detected, subsequently emerging Omicron subvariants escaped 30-60% of recently acquired natural immunity and that natural immunity lasted only one to eight months on average. We documented our analyses algorithmically and present our methods in conjunction with interactive online code notebooks and plots. We demonstrate the feasibility of integrating a flexible domain-specific syntax library with state-of-the-art packages in high performance computing, calibration, optimisation and visualisation to create an end-to-end pipeline for infectious disease modelling. We used the resulting platform to demonstrate key epidemiological characteristics of the transition from the emergency to the endemic phase of the COVID-19 pandemic.
Collapse
Affiliation(s)
- James M. Trauer
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Angus E. Hughes
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - David S. Shipman
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Michael T. Meehan
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Alec S. Henderson
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Emma S. McBryde
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Romain Ragonnet
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
2
|
Cai J, Zhang H, Zhu K, Zhu F, Wang Y, Wang S, Xie F, Zhang M, Rui L, Li S, Lin K, Xue Q, Yuan G, Wang H, Zhang Y, Fu Z, Song J, Zhang Y, Ai J, Zhang W. Risk of reinfection and severity with the predominant BA.5 Omicron subvariant China, from December 2022 to January 2023. Emerg Microbes Infect 2024; 13:2292071. [PMID: 38054806 PMCID: PMC10849001 DOI: 10.1080/22221751.2023.2292071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/03/2023] [Indexed: 12/07/2023]
Abstract
Data on reinfection in large Asian populations are limited. In this study, we aimed to evaluate the reinfection rate, disease severity, and time interval between the infections in the symptomatic and asymptomatic populations which are firstl infected with BA.2 Omicron Variant. We retrospectively included adult patients with COVID-19 discharged from four designated hospitals between 27 April 2021 and 30 November 2022, who were interviewed via telephone from 29 January to 1 March 2023. Univariable and multivariable analyses were used to explore risk factors associated with reinfection. A total of 16,558 patients were followed up, during the telephone survey of an average of 310.0 days, 1610 (9.72%) participants self-reported reinfection. The mean time range of reinfection was 257.9 days. The risks for reinfection were analysed using multivariable logistic regression. Patients with severe first infection were at higher risk for reinfection (aORs, 2.50; P < 0.001). The male (aORs,0.82; P < 0.001), the elderly (aORs, 0.44; P < 0.001), and patients with full vaccination (aORs, 0.67; P < 0.001) or booster (aORs, 0.63; P < 0.001) had the lower risk of reinfection. Patients over 60 years of age (aORs,9.02; P = 0.006) and those with ≥2 comorbidities (aORs,11.51; P = 0.016). were at higher risk for severe reinfection. The number of clinical manifestations of reinfection increases in people with severe first infection (aORs, 2.82; P = 0.023). The overall reinfection rate was 9.72%, and the reinfection rate of Omicron-to-Omicron subvariants was 9.50% at one year. The severity of Omicron-Omicron reinfection decreased. Data from our clinical study may provide clinical evidence and bolster response preparedness for future COVID-19 reinfection waves.
Collapse
Affiliation(s)
- Jianpeng Cai
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Haocheng Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Kun Zhu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Feng Zhu
- Department of Respiratory and Critical Care Medicine, Affiliated Wuxi Fifth Hospital of Jiangnan University, Wuxi Fifth People's Hospital, Wuxi, People’s Republic of China
| | - Yan Wang
- Department of Infectious Diseases, The Sixth People’s Hospital of Shenyang, Shenyang, People’s Republic of China
| | - Sen Wang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Faren Xie
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, People’s Republic of China
| | - Meng Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, People’s Republic of China
| | - Lili Rui
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, People’s Republic of China
| | - Shuhong Li
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, People’s Republic of China
| | - Ke Lin
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Quanlin Xue
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Guanmin Yuan
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Hongyu Wang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Yi Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Zhangfan Fu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Jieyu Song
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Yanliang Zhang
- Department of Infectious Diseases, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Nanjing Research Center for Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Nanjing, People’s Republic of China
| | - Jingwen Ai
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Shanghai Huashen Institute of Microbes and Infections, Shanghai, People’s Republic of China
| |
Collapse
|
3
|
Chen D, Zhang W, Xiao B, Xu B, Yang X, Deng S, Li G, Yang G, Cao J, Mei X, Luo Q, Huang P, Sun X, Su J, Zhong N, Zhao Z, Wang Z. Effect of wild-type vaccine doses on BA.5 hybrid immunity, disease severity, and XBB reinfection risk. J Virol 2024:e0128524. [PMID: 39499071 DOI: 10.1128/jvi.01285-24] [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: 07/22/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Vaccination against the wild-type (WT) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus did not produce detectable levels of neutralizing antibodies (NAbs) against the BA.5 strain before it emerged. However, coronavirus disease-2019 (COVID-19) severity varied highly between unvaccinated, partially vaccinated, and fully vaccinated individuals, for unknown reasons. We assessed the severity of BA.5 infection and the risk of XBB strain reinfection and measured serum levels of NAbs against WT, BA.5, and XBB.1.9.1 SARS-CoV-2 strains at varying time points in 1,373 individuals who received zero, one, two, or three WT vaccine doses. We found that two to three WT doses significantly increased WT and BA.5 NAb levels and reduced the incidence of COVID-19-associated pneumonia upon BA.5 strain infection compared to zero to one dose. Regarding XBB reinfection, those who received two to three doses and were infected with the BA.5 variant exhibited a significantly lower reinfection risk compared to those who received zero to one dose. RNA analysis revealed that the differentially expressed genes between the two to three dose and unvaccinated groups were enriched in B cell activation, cytokine-cytokine receptor interaction, complement, and monocyte activation functions-indicating that vaccination increased the antibody response and reduced inflammation. Our results suggest that multiple antigen exposures to either matched or unmatched SARS-COV-2 variants, through vaccination or infection, may be necessary to achieve significant immune imprinting.IMPORTANCEThe administration of coronavirus disease-2019 (COVID-19) vaccines that do not perfectly match the viral strains that individuals become infected with has been found to impact the resultant illness severity-although the precise mechanism underlying this phenomenon remains unclear. We assessed viral clearance, as well as serum levels of inflammatory cytokines and neutralizing antibodies (NAbs) against wild-type, BA.5, and XBB.1.9.1 variants of the severe acute respiratory syndrome coronavirus 2 among individuals who received varying doses of such strain-mismatched vaccines. Notably, vaccination with ≥2 doses of strain-mismatched COVID-19 vaccines appeared to stimulate the production of specific NAbs during infection with new variants, as well as attenuate the inflammatory response and enhance viral clearance. Such vaccination regimens can also reduce the risk of reinfection. These findings may be important for guiding the development of future COVID-19 vaccination strategies that target both matched and mismatched viral variants.
Collapse
Affiliation(s)
- Daxiang 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Weihong Zhang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyun Yang
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Shidong Deng
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Guichang Li
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Gang Yang
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Jinpeng Cao
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Xinyue Mei
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Qi Luo
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Peiyu Huang
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Xi Sun
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Jie Su
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Nanshan Zhong
- 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| | - Zhuxiang Zhao
- Department of Infectious Disease, Respiratory and Critical Care Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhongfang 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 Medical University, Guangzhou, Guangdong, China
- Hetao Institute of Guangzhou National Laboratory, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
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
|
5
|
Yang Y, Hu J, Deng H, Chen D, Wu G, Xing H, Liu Y, Li S, Yan Y, Tang N, Zhao Y. Maternal Preconception COVID-19 Vaccination and Its Protective Effect on Infants after a Breakthrough Infection during Pregnancy. Vaccines (Basel) 2024; 12:1132. [PMID: 39460299 PMCID: PMC11511423 DOI: 10.3390/vaccines12101132] [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: 09/02/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND AND AIMS The transplacental vertical transfer of maternal antibodies was determined to be a crucial factor in conferring protective immunity to infants following delivery, and this study aimed to evaluate the protective effect of maternal preconception COVID-19 vaccination on infants. METHODS A prospective cohort study was conducted at the National Clinical Medical Research Center for Child Health and Diseases in Chongqing, China, spanning from July 2022 to April 2023. The study included infants from mothers with a preconception COVID-19 vaccination and (or) a SARS-CoV-2 infection during pregnancy. Titers of SARS-CoV-2 immunoglobulin G (IgG) and cross-neutralizing activity against SARS-CoV-2 variants were detected. RESULTS In this cohort study comprising 158 infants, it was observed that infants born to mothers who experienced a pregnancy-related breakthrough infection following a preconception vaccination had the highest titers of SARS-CoV-2 IgG and cross-neutralizing antibody activity against different variants compared to those with either of these factors alone. The transplacental vertical transmission of anti-SARS-CoV-2 antibodies decreased significantly with increasing age, from 3.16 ODs at birth to 2.29 ODs at two months, and persisted for approximately four months after birth. The predominant subclass of passively transmitted antibodies via the placenta was found to be IgG1, and a positive correlation was observed between the titers of SARS-CoV-2 IgG and IgG1 (R = 0.59, p < 0.001; Slope: 0.49 ± 0.070, p < 0.001). CONCLUSIONS Maternal preconception COVID-19 vaccination represents a promising immunological strategy for conferring postnatal protection to infants, especially during the period of heightened risk of exposure to SARS-CoV-2 infection. It is imperative to underscore the significance of vaccination for women who are preparing to become pregnant or are pregnant, and concerted efforts must be made to promote vaccination among eligible women.
Collapse
Affiliation(s)
- Yuting Yang
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children’s Hospital of Chongqing Medical University, Chongqing 400015, China; (Y.Y.); (D.C.); (G.W.); (H.X.)
| | - Jie Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; (J.H.); (H.D.); (Y.L.); (S.L.); (Y.Y.)
- Department of Laboratory Medicine, Bishan Hospital of Chongqing Medical University, Chongqing 402760, China
| | - Haijun Deng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; (J.H.); (H.D.); (Y.L.); (S.L.); (Y.Y.)
| | - Dapeng Chen
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children’s Hospital of Chongqing Medical University, Chongqing 400015, China; (Y.Y.); (D.C.); (G.W.); (H.X.)
| | - Guojin Wu
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children’s Hospital of Chongqing Medical University, Chongqing 400015, China; (Y.Y.); (D.C.); (G.W.); (H.X.)
| | - Huiwu Xing
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children’s Hospital of Chongqing Medical University, Chongqing 400015, China; (Y.Y.); (D.C.); (G.W.); (H.X.)
| | - Yuanyuan Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; (J.H.); (H.D.); (Y.L.); (S.L.); (Y.Y.)
| | - Shan Li
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; (J.H.); (H.D.); (Y.L.); (S.L.); (Y.Y.)
| | - Yihan Yan
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; (J.H.); (H.D.); (Y.L.); (S.L.); (Y.Y.)
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; (J.H.); (H.D.); (Y.L.); (S.L.); (Y.Y.)
| | - Yao Zhao
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children’s Hospital of Chongqing Medical University, Chongqing 400015, China; (Y.Y.); (D.C.); (G.W.); (H.X.)
| |
Collapse
|
6
|
García-Pérez J, Borobia AM, Pérez-Olmeda M, Portolés A, Castaño L, Campins-Artí M, Bertrán MJ, Bermejo M, Arribas JR, López A, Ascaso-Del-Rio A, Arana-Arri E, Fuentes Camps I, Vilella A, Cascajero A, García-Morales MT, Castillo de la Osa M, Pérez Ingidua C, Lora D, Jiménez-Santana P, Pino-Rosa S, Gómez de la Cámara A, De La Torre-Tarazona E, Calonge E, Cruces R, Belda-Iniesta C, Alcamí J, Frías J, Carcas AJ, Díez-Fuertes F. Immunogenicity of a third dose with mRNA-vaccines in the ChAdOx1-S/BNT162b2 vaccination regimen against SARS-CoV-2 variants. iScience 2024; 27:110728. [PMID: 39286494 PMCID: PMC11404211 DOI: 10.1016/j.isci.2024.110728] [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: 03/04/2024] [Revised: 06/28/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
CombiVacS study has demonstrated a strong immune response of the heterologous ChAdOx1-S/BNT162b2 vaccine combination. The primary outcomes of the study were to assess the humoral immune response against SARS-CoV-2, 28 days after a third dose of a mRNA vaccine, in subjects that received a previous prime-boost scheme with ChAdOx1-S/BNT162b2. Secondary outcomes extended the study to 3 and 6 months. The third vaccine dose of mRNA-1273 in naive participants previously vaccinated with ChAdOx1-S/BNT162b2 regimen reached higher neutralizing antibodies titers against the variants of concern Delta and BA.1 lineage of Omicron compared with those receiving a third dose of BNT162b2 at day 28. These differences between BNT162b2 and mRNA-1273 arms were observed against the ancestral variant G614 at day 90. Suboptimal neutralizing response was observed against BQ.1.1, XBB.1.5/XBB.1.9, and JN.1 in a relevant proportion of individuals 180 days after the third dose, even after asymptomatic Omicron breakthrough infections. EudraCT (2021-001978-37); ClinicalTrials.gov (NCT04860739).
Collapse
Affiliation(s)
- Javier García-Pérez
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Alberto M Borobia
- Servicio de Farmacología Clínica, Hospital Universitario La Paz, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, IdiPAZ, 28046 Madrid, Spain
- Servicio de Medicina Interna, Departamento de Medicina, Facultad de Medicina, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, 28046 Madrid, Spain
| | - Mayte Pérez-Olmeda
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Antonio Portolés
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - Luis Castaño
- Cruces University Hospital, Bio-Bizkaia, UPV/EHU, CIBERDEM/CIBERER, Endo-ERN, Barakaldo, 48903 Vizcaya, Spain
| | - Magdalena Campins-Artí
- Servicio de Medicina Preventiva y Epidemiología, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - María Jesús Bertrán
- Servicio de Medicina Preventiva y Epidemiología, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Mercedes Bermejo
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - José Ramón Arribas
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Servicio de Medicina Interna, Departamento de Medicina, Facultad de Medicina, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, 28046 Madrid, Spain
| | - Andrea López
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Ana Ascaso-Del-Rio
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | - Eunate Arana-Arri
- Cruces University Hospital, Bio-Bizkaia, UPV/EHU, CIBERDEM/CIBERER, Endo-ERN, Barakaldo, 48903 Vizcaya, Spain
| | - Inmaculada Fuentes Camps
- Unidad de Soporte a la Investigación Clínica, Vall d'Hebron Institut de Recerca, Servicio de Farmacología Clínica, Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Anna Vilella
- Servicio de Medicina Preventiva y Epidemiología, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Almudena Cascajero
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | | | - María Castillo de la Osa
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Carla Pérez Ingidua
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | - David Lora
- Spanish Clinical Research Network - SCReN - ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario 12 de octubre, 28041 Madrid, Spain
- Facultad de Estudios Estadísticos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Paloma Jiménez-Santana
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Silvia Pino-Rosa
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Agustín Gómez de la Cámara
- Instituto de Investigación Sanitaria del Hospital Universitario 12 de octubre, 28041 Madrid, Spain
- Facultad de Estudios Estadísticos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Erick De La Torre-Tarazona
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Esther Calonge
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Raquel Cruces
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | | | - José Alcamí
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Jesús Frías
- Servicio de Farmacología Clínica, Hospital Universitario La Paz, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, IdiPAZ, 28046 Madrid, Spain
| | - Antonio J Carcas
- Servicio de Farmacología Clínica, Hospital Universitario La Paz, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, IdiPAZ, 28046 Madrid, Spain
| | - Francisco Díez-Fuertes
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| |
Collapse
|
7
|
See KC. Enhancing COVID-19 Vaccination Awareness and Uptake in the Post-PHEIC Era: A Narrative Review of Physician-Level and System-Level Strategies. Vaccines (Basel) 2024; 12:1038. [PMID: 39340068 PMCID: PMC11435511 DOI: 10.3390/vaccines12091038] [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: 07/28/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Following the World Health Organization's declaration that the COVID-19 pandemic is no longer a public health emergency of international concern (PHEIC), COVID-19 remains an ongoing threat to human health and healthcare systems. Vaccination plays a crucial role in reducing the disease's incidence, mitigating its severity, and limiting transmission, contributing to long-term public health resilience. However, incomplete vaccination coverage and vaccine hesitancy exist. This narrative review investigates strategies at the system and physician levels aimed at sustaining awareness and uptake of COVID-19 vaccination in a post-PHEIC era. Through an examination of the existing literature, this review explores the effectiveness of diverse approaches utilized by healthcare systems and individual providers. These approaches address every component of the 5C model of vaccine hesitancy: confidence, complacency, constraints/convenience, calculation, and collective responsibility. Physician-level approaches include appropriate message framing, persuasive communication containing safety and personal/social benefit information, sharing of personal stories, creating a safe space for discussion, harnessing co-administration with annual influenza vaccines, and use of decision aids and visual messages. System-level approaches include messaging, mass media for health communication, on-site vaccine availability, pharmacist delivery, healthcare protocol integration, incentives, and chatbot use.
Collapse
Affiliation(s)
- Kay Choong See
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, Singapore 119074, Singapore
| |
Collapse
|
8
|
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
|
9
|
Poh XY, Lee IR, Tan CW, Chavatte JM, Fong SW, Goh YS, Rouers A, Wong N, Torres-Ruesta A, Mah SYY, Yeoh AYY, Gandhi M, Rahman N, Chin YQ, Lim JJ, Yoong TJK, Rao S, Chia PY, Ong SWX, Lee TH, Sadarangani SP, Lin RJH, Lim DRX, Chia W, Renia L, Ren EC, Lin RTP, Lye DC, Wang LF, Ng LFP, Young BE. First SARS-CoV-2 Omicron infection as an effective immune booster among mRNA vaccinated individuals: final results from the first phase of the PRIBIVAC randomised clinical trial. EBioMedicine 2024; 107:105275. [PMID: 39137572 PMCID: PMC11367514 DOI: 10.1016/j.ebiom.2024.105275] [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: 03/01/2024] [Revised: 07/14/2024] [Accepted: 07/27/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Understanding how SARS-CoV-2 breakthrough infections impacts the breadth of immune responses against existing and pre-emergent SARS-CoV-2 strains is needed to develop an evidence-based long-term immunisation strategy. METHODS We performed a randomised, controlled trial to assess the immunogenicity of homologous (BNT162b2) versus heterologous (mRNA-1273) booster vaccination in 100 BNT162b2-vaccinated infection-naïve individuals enrolled from October 2021. Post hoc analysis was performed to assess the impact of SARS-CoV-2 infection on humoral and cellular immune responses against wild-type SARS-CoV-2 and/or Omicron subvariants. FINDINGS 93 participants completed the study at day 360. 71% (66/93) of participants reported first SARS-CoV-2 Omicron infection by the end of the study with similar proportions of infections between homologous and heterologous booster groups (72.3% [34/47] vs 69.6% [32/46]; p = 0.82). Mean wildtype SARS-CoV-2 anti-S-RBD antibody level was significantly higher in heterologous booster group compared with homologous group at day 180 (14,588 IU/mL; 95% CI, 10,186-20,893 vs 7447 IU/mL; 4646-11,912; p = 0.025). Participants who experienced breakthrough infections during the Omicron BA.1/2 wave had significantly higher anti-S-RBD antibody levels against wildtype SARS-CoV-2 and antibody neutralisation against BA.1 and pre-emergent BA.5 compared with infection-naïve participants. Regardless of hybrid immunity status, wildtype SARS-CoV-2 anti-S-RBD antibody level declined significantly after six months post-booster or post-SARS-CoV-2 infection. INTERPRETATION Booster vaccination with mRNA-1273 was associated with significantly higher antibody levels compared with BNT162b2. Antibody responses are narrower and decline faster among uninfected, vaccinated individuals. Boosters may be more effective if administered shortly before infection outbreaks and at least six months after last infection or booster. FUNDING Singapore NMRC, USFDA, MRC.
Collapse
Affiliation(s)
| | - I Russel Lee
- National Centre for Infectious Diseases, Singapore
| | - Chee Wah Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jean-Marc Chavatte
- National Centre for Infectious Diseases, Singapore; National Public Health Laboratory, Singapore
| | - Siew Wai Fong
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), Singapore, 138648, Singapore
| | - Yun Shan Goh
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), Singapore, 138648, Singapore
| | - Angeline Rouers
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), Singapore, 138648, Singapore
| | - Nathan Wong
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), Singapore, 138648, Singapore
| | - Anthony Torres-Ruesta
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), Singapore, 138648, Singapore
| | - Shirley Y Y Mah
- Emerging Infectious Diseases Programme, Duke-NUS Medical School, Singapore
| | - Aileen Y Y Yeoh
- Emerging Infectious Diseases Programme, Duke-NUS Medical School, Singapore
| | - Mihir Gandhi
- Biostatistics, Singapore Clinical Research Institute, Singapore; Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Nabilah Rahman
- Biostatistics, Singapore Clinical Research Institute, Singapore; Saw Swee Hock School of Public Health, Singapore
| | - Yi Qing Chin
- National Centre for Infectious Diseases, Singapore
| | | | | | - Suma Rao
- National Centre for Infectious Diseases, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Po Ying Chia
- National Centre for Infectious Diseases, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Sean W X Ong
- National Centre for Infectious Diseases, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Tau Hong Lee
- National Centre for Infectious Diseases, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Sapna P Sadarangani
- National Centre for Infectious Diseases, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Ray J H Lin
- National Centre for Infectious Diseases, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Daniel R X Lim
- National Centre for Infectious Diseases, Singapore; National Public Health Laboratory, Singapore
| | - Wanni Chia
- Emerging Infectious Diseases Programme, Duke-NUS Medical School, Singapore
| | - Laurent Renia
- A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), Singapore, 138648, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ee Chee Ren
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore Immunology Network, Singapore
| | - Raymond T P Lin
- National Centre for Infectious Diseases, Singapore; National Public Health Laboratory, Singapore
| | - David C Lye
- National Centre for Infectious Diseases, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Lin-Fa Wang
- Emerging Infectious Diseases Programme, Duke-NUS Medical School, Singapore
| | - Lisa F P Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; A∗STAR Infectious Diseases Labs (A∗STAR ID Labs), Agency for Science, Technology and Research (A∗STAR), Singapore, 138648, Singapore.
| | - Barnaby E Young
- National Centre for Infectious Diseases, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.
| |
Collapse
|
10
|
Zheng H, Wu S, Chen W, Cai S, Zhan M, Chen C, Lin J, Xie Z, Ou J, Ye W. Meta-analysis of hybrid immunity to mitigate the risk of Omicron variant reinfection. Front Public Health 2024; 12:1457266. [PMID: 39253287 PMCID: PMC11381385 DOI: 10.3389/fpubh.2024.1457266] [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: 07/08/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Background Hybrid immunity (a combination of natural and vaccine-induced immunity) provides additional immune protection against the coronavirus disease 2019 (COVID-19) reinfection. Today, people are commonly infected and vaccinated; hence, hybrid immunity is the norm. However, the mitigation of the risk of Omicron variant reinfection by hybrid immunity and the durability of its protection remain uncertain. This meta-analysis aims to explore hybrid immunity to mitigate the risk of Omicron variant reinfection and its protective durability to provide a new evidence-based basis for the development and optimization of immunization strategies and improve the public's awareness and participation in COVID-19 vaccination, especially in vulnerable and at-risk populations. Methods Embase, PubMed, Web of Science, Chinese National Knowledge Infrastructure, and Wanfang databases were searched for publicly available literature up to 10 June 2024. Two researchers independently completed the data extraction and risk of bias assessment and cross-checked each other. The Newcastle-Ottawa Scale assessed the risk of bias in included cohort and case-control studies, while criteria recommended by the Agency for Health Care Research and Quality (AHRQ) evaluated cross-sectional studies. The extracted data were synthesized in an Excel spreadsheet according to the predefined items to be collected. The outcome was Omicron variant reinfection, reported as an Odds Ratio (OR) with its 95% confidence interval (CI) and Protective Effectiveness (PE) with 95% CI. The data were pooled using a random- or fixed-effects model based on the I2 test. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. Results Thirty-three articles were included. Compared with the natural immunity group, the hybrid immunity (booster vaccination) group had the highest level of mitigation in the risk of reinfection (OR = 0.43, 95% CI:0.34-0.56), followed by the complete vaccination group (OR = 0.58, 95% CI:0.45-0.74), and lastly the incomplete vaccination group (OR = 0.64, 95% CI:0.44-0.93). Compared with the complete vaccination-only group, the hybrid immunity (complete vaccination) group mitigated the risk of reinfection by 65% (OR = 0.35, 95% CI:0.27-0.46), and the hybrid immunity (booster vaccination) group mitigated the risk of reinfection by an additional 29% (OR = 0.71, 95% CI:0.61-0.84) compared with the hybrid immunity (complete vaccination) group. The effectiveness of hybrid immunity (incomplete vaccination) in mitigating the risk of reinfection was 37.88% (95% CI, 28.88-46.89%) within 270-364 days, and decreased to 33.23%% (95% CI, 23.80-42.66%) within 365-639 days; whereas, the effectiveness after complete vaccination was 54.36% (95% CI, 50.82-57.90%) within 270-364 days, and the effectiveness of booster vaccination was 73.49% (95% CI, 68.95-78.04%) within 90-119 days. Conclusion Hybrid immunity was significantly more protective than natural or vaccination-induced immunity, and booster doses were associated with enhanced protection against Omicron. Although its protective effects waned over time, vaccination remains a crucial measure for controlling COVID-19. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier, CRD42024539682.
Collapse
Affiliation(s)
- Huiling Zheng
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
- School of Public Health, Fujian Medical University, Fuzhou, China
| | - Shenggen Wu
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Wu Chen
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Shaojian Cai
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Meirong Zhan
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Cailin Chen
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Jiawei Lin
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Zhonghang Xie
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Jianming Ou
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| | - Wenjing Ye
- Institute of Emergency Response and Epidemic Management, Fujian Provincial Center for Disease Control and Prevention, Fuzhou, China
| |
Collapse
|
11
|
Kung YA, Chuang CH, Chen YC, Yang HP, Li HC, Chen CL, Janapatla RP, Chen CJ, Shih SR, Chiu CH. Worldwide SARS-CoV-2 Omicron variant infection: Emerging sub-variants and future vaccination perspectives. J Formos Med Assoc 2024:S0929-6646(24)00389-9. [PMID: 39179492 DOI: 10.1016/j.jfma.2024.08.021] [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: 02/06/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has sparked widespread concern globally, particularly with the Omicron variant and its sub-lineages emerging as the predominant cause of infection for nearly two years. Taiwan's successful containment of COVID-19, underscored by broad vaccine coverage, the utilization of anti-viral therapeutics, and timely response strategies, has resulted in reduced excess mortality. Moreover, there is a crucial need for a phased exit strategy, balancing efforts to curtail disease transmission with the mitigation of socioeconomic impacts from rigorous measures. In this review, we examined the evolution and the epidemiological landscape of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron sub-variants in Taiwan as well as other countries of the world. We also critically evaluated the effectiveness of COVID-19 vaccines against various SARS-CoV-2 variants. Additionally, we addressed the advantages of heterologous immunization strategies, fluctuations in neutralizing antibody titers, and complexities in establishing protective correlates among swiftly mutating viral variants.
Collapse
Affiliation(s)
- Yu-An Kung
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Hsien Chuang
- Department of Pediatrics, St. Paul's Hospital, Taoyuan, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan; School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Yi-Ching Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hsin-Ping Yang
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsin-Chieh Li
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chyi-Liang Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | - Chin-Jung Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
12
|
Barroso-Arévalo S, Sánchez-Morales L, Porras N, Díaz-Frutos M, Barasona JA, Isla J, López D, Gortázar C, Domínguez L, Sánchez-Vizcaíno JM. Comparative SARS-CoV-2 Omicron BA.5 variant and D614G-Wuhan strain infections in ferrets: insights into attenuation and disease progression during subclinical to mild COVID-19. Front Vet Sci 2024; 11:1435464. [PMID: 39211479 PMCID: PMC11358085 DOI: 10.3389/fvets.2024.1435464] [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: 05/20/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction As the SARS-CoV-2 virus continues to evolve and new variants emerge, it becomes crucial to understand the comparative pathological and immunological responses elicited by different strains. This study focuses on the original Wuhan strain and the Omicron variant, which have demonstrated significant differences in clinical outcomes and immune responses. Methods We employed ferrets as an experimental model to assess the D614G variant (a derivative of the Wuhan strain) and the Omicron BA.5 variant. Each variant was inoculated into separate groups of ferrets to compare disease severity, viral dissemination, and immune responses. Results The D614G variant induced more severe disease and greater viral spread than the Omicron variant. Notably, ferrets infected with the D614G variant exhibited a robust neutralizing antibody response, whereas those infected with the Omicron variant failed to produce a detectable neutralizing antibody response. Despite the clearance of the virus from nearly all tissues by 7 days post-infection, an increase in pathological lesions was observed from 14 to 21 days, particularly in those infected with the D614G variant, suggesting a sustained immune response even after viral clearance. Discussion These findings underscore the adaptability of SARS-CoV-2 and illuminate how susceptibility and clinical manifestations vary across different strains and species. The results emphasize the necessity of considering both the direct effects of viral infection and the indirect, often prolonged, impacts of the immune response in evaluating the outcomes of SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Sandra Barroso-Arévalo
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Lidia Sánchez-Morales
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Néstor Porras
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Marta Díaz-Frutos
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Jose A. Barasona
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Débora López
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Christian Gortázar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Lucas Domínguez
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| | - Jose M. Sánchez-Vizcaíno
- Department of Animal Health, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
- VISAVET Health Surveillance Centre, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
13
|
Xiong Y, Wang C, Zhang Y. Interacting particle models on the impact of spatially heterogeneous human behavioral factors on dynamics of infectious diseases. PLoS Comput Biol 2024; 20:e1012345. [PMID: 39116182 PMCID: PMC11335169 DOI: 10.1371/journal.pcbi.1012345] [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: 01/30/2024] [Revised: 08/20/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Human behaviors have non-negligible impacts on spread of contagious disease. For instance, large-scale gathering and high mobility of population could lead to accelerated disease transmission, while public behavioral changes in response to pandemics may effectively reduce contacts and suppress the peak of the outbreak. In order to understand how spatial characteristics like population mobility and clustering interplay with epidemic outbreaks, we formulate a stochastic-statistical environment-epidemic dynamic system (SEEDS) via an agent-based biased random walk model on a two-dimensional lattice. The "popularity" and "awareness" variables are taken into consideration to capture human natural and preventive behavioral factors, which are assumed to guide and bias agent movement in a combined way. It is found that the presence of the spatial heterogeneity, like social influence locality and spatial clustering induced by self-aggregation, potentially suppresses the contacts between agents and consequently flats the epidemic curve. Surprisedly, disease responses might not necessarily reduce the susceptibility of informed individuals and even aggravate disease outbreak if each individual responds independently upon their awareness. The disease control is achieved effectively only if there are coordinated public-health interventions and public compliance to these measures. Therefore, our model may be useful for quantitative evaluations of a variety of public-health policies.
Collapse
Affiliation(s)
- Yunfeng Xiong
- School of Mathematical Sciences, Beijing Normal University, Beijing, China
| | - Chuntian Wang
- Department of Mathematics, The University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Yuan Zhang
- Center for Applied Statistics and School of Statistics, Renmin University of China, Bejing, China
| |
Collapse
|
14
|
Zhang J. Immune responses in COVID-19 patients: Insights into cytokine storms and adaptive immunity kinetics. Heliyon 2024; 10:e34577. [PMID: 39149061 PMCID: PMC11325674 DOI: 10.1016/j.heliyon.2024.e34577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024] Open
Abstract
SARS-CoV-2 infection can trigger cytokine storm in some patients, which characterized by an excessive production of cytokines and chemical mediators. This hyperactive immune response may cause significant tissue damage and multiple organ failure (MOF). The severity of COVID-19 correlates with the intensity of cytokine storm, involving elements such as IFN, NF-κB, IL-6, HMGB1, etc. It is imperative to rapidly engage adaptive immunity to effectively control the disease progression. CD4+ T cells facilitate an immune response by improving B cells in the production of neutralizing antibodies and activating CD8+ T cells, which are instrumental in eradicating virus-infected cells. Meanwhile, antibodies from B cells can neutralize virus, obstructing further infection of host cells. In individuals who have recovered from the disease, virus-specific antibodies and memory T cells were observed, which could confer a level of protection, reducing the likelihood of re-infection or attenuating severity. This paper discussed the roles of macrophages, IFN, IL-6 and HMGB1 in cytokine release syndrome (CRS), the intricacies of adaptive immunity, and the persistence of immune memory, all of which are critical for the prevention and therapeutic strategies against COVID-19.
Collapse
Affiliation(s)
- Junguo Zhang
- Pulmonology Department, Fengdu General Hospital, Chongqing, 408200, China
| |
Collapse
|
15
|
Lee J, Naoe Y, Bang U, Nakagama Y, Saito A, Kido Y, Hotta A. Neutralization sensitivity of SARS-CoV-2 Omicron variants FL.1 and GE.1 by therapeutic antibodies and XBB sera. Virology 2024; 595:110067. [PMID: 38653156 DOI: 10.1016/j.virol.2024.110067] [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: 05/18/2023] [Revised: 02/22/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
Two SARS-CoV-2 XBB sub-variants, FL.1 and GE.1, have been increasing in prevalence worldwide, but limited information is available about their ability to evade the immune system. FL.1 and GE.1 are emerging Omicron XBB variants possessing additional mutations in the spike RBD raising concerns of increased neutralization escape. In this study, we assessed the neutralizing ability of eleven FDA-approved monoclonal antibody combinations against different Omicron variants, including BA.2.75, BA.2.76, BA.4/5, XBB.1.5, and CH.1.1. Among the eleven antibodies, Sotrovimab was the only antibody to show broad neutralization ability against XBB.1.5. However, Sotrovimab showed attenuated neutralization efficiency against recently emerging XBB sub-lineages EG.5, FL.1, and GE.1 compared to XBB.1.5. Additionally, XBB.1.5 seropositive convalescent sera displayed lower neutralization activity against EG.5, FL.1, and GE.1. Overall, our findings present enhanced immune evasion capacity of emerging XBB variants and emphasize the importance of continued monitoring of novel variants.
Collapse
Affiliation(s)
- Joseph Lee
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Youichi Naoe
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Uikyu Bang
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan
| | - Yu Nakagama
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akatsuki Saito
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan; Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Yasutoshi Kido
- Department of Virology & Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, 606-8507, Japan.
| |
Collapse
|
16
|
Lee B, Bae GE, Jeong IH, Kim JH, Kwon MJ, Kim J, Kim B, Lee JW, Nam JH, Huh HJ, Kang ES. Age-Related Differences in Neutralizing Antibody Responses against SARS-CoV-2 Delta and Omicron Variants in 151 SARS-CoV-2-Naïve Metropolitan Residents Boosted with BNT162b2. J Appl Lab Med 2024; 9:741-751. [PMID: 38531067 DOI: 10.1093/jalm/jfae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/09/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Although age negatively correlates with vaccine-induced immune responses, whether the vaccine-induced neutralizing effect against variants of concern (VOCs) substantially differs across age remains relatively poorly explored. In addition, the utility of commercial binding assays developed with the wild-type SARS-CoV-2 for predicting the neutralizing effect against VOCs should be revalidated. METHODS We analyzed 151 triple-vaccinated SARS-CoV-2-naïve individuals boosted with BNT162b2 (Pfizer-BioNTech). The study population was divided into young adults (age < 30), middle-aged adults (30 ≤ age < 60), and older adults (age ≥ 60). The plaque reduction neutralization test (PRNT) titers against Delta (B.1.617.2) and Omicron (B.1.1.529) variants were compared across age. Antibody titers measured with commercial binding assays were compared with PRNT titers. RESULTS Age-related decline in neutralizing titers was observed for both Delta and Omicron variants. Neutralizing titers for Omicron were lower than those against Delta in all ages. The multiple linear regression model demonstrated that duration from third dose to sample collection and vaccine types were also significant factors affecting vaccine-induced immunity along with age. The correlation between commercial binding assays and PRNT was acceptable for all age groups with the Delta variant, but relatively poor for middle-aged and older adults with the Omicron variant due to low titers. CONCLUSIONS This study provides insights into the age-related dynamics of vaccine-induced immunity against SARS-CoV-2 VOCs, corroborating the need for age-specific vaccination strategies in the endemic era where new variants continue to evolve. Moreover, commercial binding assays should be used cautiously when estimating neutralizing titers against VOCs, particularly Omicron.
Collapse
Affiliation(s)
- Beomki Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Go Eun Bae
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - In Hwa Jeong
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Laboratory Medicine, Dong-A University Hospital, Busan, Republic of Korea
| | - Jong-Hun Kim
- Department of Social and Preventive Medicine, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, Republic of Korea
| | - Min-Jung Kwon
- Department of Laboratory Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jayoung Kim
- Department of Laboratory Medicine, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, Republic of Korea
| | - Byoungguk Kim
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - June-Woo Lee
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Jeong-Hyun Nam
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Hee Jin Huh
- Department of Laboratory Medicine, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Eun-Suk Kang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Guo X, Cheng Z, Li J, Song Y, Zheng H, Wang Y, Ma C, Feng Z. Protection of Omicron Sub-Lineage Infection Against Reinfection with Another Omicron Sub-Lineage: Systematic Review, Meta-Analysis, and Meta-Regression - Worldwide, 2022-2023. China CDC Wkly 2024; 6:624-628. [PMID: 38966308 PMCID: PMC11219298 DOI: 10.46234/ccdcw2024.103] [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: 12/13/2023] [Accepted: 05/27/2024] [Indexed: 07/06/2024] Open
Abstract
What is already known about this topic? Both the decline in immunity over time and the evolution of the virus play a role in the level of protection offered by a prior infection. What is added by this report? Point estimates indicated variations in protection levels based on the initial infecting variant and the reinfecting variant. There was a consistent correlation between real-world protection, antigenic distance, and humoral immunity levels. Specifically, shorter antigenic distances and higher humoral immunity levels corresponded to enhanced real-world protection. What are the implications for public health practice? Our findings suggest that virological and immunological studies could help identify and assess the epidemic risk posed by new variants before they become dominant. Prompt incorporation of the latest variants into the antigen components of the coronavirus disease 2019 (COVID-19) vaccines can significantly contribute to effective epidemic prevention and control measures.
Collapse
Affiliation(s)
- Xu Guo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zuyao Cheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Junhong Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yudan Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Zheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yamin Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chao Ma
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zijian Feng
- Chinese Preventive Medicine Association, Beijing, China
| |
Collapse
|
18
|
Chivu CD, Crăciun MD, Pițigoi D, Aramă V, Luminos ML, Jugulete G, Nițescu VG, Lescaie A, Apostolescu CG, Streinu Cercel A. Hybrid Immunity and the Incidence of SARS-CoV-2 Reinfections during the Omicron Era in Frontline Healthcare Workers. Vaccines (Basel) 2024; 12:682. [PMID: 38932411 PMCID: PMC11209586 DOI: 10.3390/vaccines12060682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
During the coronavirus disease (COVID-19) pandemic healthcare workers (HCWs) acquired immunity by vaccination or exposure to multiple variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our study is a comparative analysis between subgroups of HCWs constructed based on the number of SARS-CoV-2 infections, vaccination, and the dominant variant of SARS-CoV-2 in the population. We collected and analyzed data using the χ2 test and density incidence of reinfections in Microsoft Excel for Mac, Version 16.84, and MedCalc®, 22.026. Of the 829 HCWs, 70.1% (581) had only one SARS-CoV-2 infection and 29.9% (248) had two infections. Of the subjects with two infections, 77.4% (192) worked in high-risk departments and 93.2% (231) of the second infections were registered during Omicron dominance. The density incidence of reinfections was higher in HCWs vaccinated with the primary schedule than those vaccinated with the first booster, and the incidence ratio was 2.8 (95% CI: 1.2; 6.7). The probability of reinfection was five times lower (95% CI: 2.9; 9.2) in HCWs vaccinated with the primary schedule if the first infection was acquired during Omicron dominance. The subjects vaccinated with the first booster had a density incidence of reinfection three times lower (95% CI: 1.9; 5.8) if the first infection was during Omicron. The incidence ratio in subgroups constructed based on characteristics such as gender, age group, job category, and department also registered significant differences in density incidence. The history of SARS-CoV-2 infection by variant is important when interpreting and understanding public health data and the results of studies related to vaccine efficacy for hybrid immunity subgroup populations.
Collapse
Affiliation(s)
- Carmen-Daniela Chivu
- Department of Epidemiology 1, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-D.C.); (D.P.)
- Emergency Clinical Hospital for Children “Grigore Alexandrescu”, 011743 Bucharest, Romania; (V.G.N.); (A.L.)
| | - Maria-Dorina Crăciun
- Department of Epidemiology 1, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-D.C.); (D.P.)
- Emergency Clinical Hospital for Children “Grigore Alexandrescu”, 011743 Bucharest, Romania; (V.G.N.); (A.L.)
| | - Daniela Pițigoi
- Department of Epidemiology 1, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.-D.C.); (D.P.)
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania; (V.A.); (M.L.L.); (G.J.); (C.G.A.); (A.S.C.)
| | - Victoria Aramă
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania; (V.A.); (M.L.L.); (G.J.); (C.G.A.); (A.S.C.)
- Department of Infectious Diseases 1, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Monica Luminița Luminos
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania; (V.A.); (M.L.L.); (G.J.); (C.G.A.); (A.S.C.)
- Department of Infectious Diseases 3, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Gheorghiță Jugulete
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania; (V.A.); (M.L.L.); (G.J.); (C.G.A.); (A.S.C.)
- Department of Infectious Diseases 3, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Viorela Gabriela Nițescu
- Emergency Clinical Hospital for Children “Grigore Alexandrescu”, 011743 Bucharest, Romania; (V.G.N.); (A.L.)
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andreea Lescaie
- Emergency Clinical Hospital for Children “Grigore Alexandrescu”, 011743 Bucharest, Romania; (V.G.N.); (A.L.)
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Cătălin Gabriel Apostolescu
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania; (V.A.); (M.L.L.); (G.J.); (C.G.A.); (A.S.C.)
- Department of Infectious Diseases 1, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Adrian Streinu Cercel
- National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania; (V.A.); (M.L.L.); (G.J.); (C.G.A.); (A.S.C.)
- Department of Infectious Diseases 1, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
19
|
Liang CY, Raju S, Liu Z, Li Y, Asthagiri Arunkumar G, Case JB, Scheaffer SM, Zost SJ, Acreman CM, Gagne M, Andrew SF, Carvalho Dos Anjos DC, Foulds KE, McLellan JS, Crowe JE, Douek DC, Whelan SPJ, Elbashir SM, Edwards DK, Diamond MS. Imprinting of serum neutralizing antibodies by Wuhan-1 mRNA vaccines. Nature 2024; 630:950-960. [PMID: 38749479 PMCID: PMC11419699 DOI: 10.1038/s41586-024-07539-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/08/2024] [Indexed: 06/21/2024]
Abstract
Immune imprinting is a phenomenon in which prior antigenic experiences influence responses to subsequent infection or vaccination1,2. The effects of immune imprinting on serum antibody responses after boosting with variant-matched SARS-CoV-2 vaccines remain uncertain. Here we characterized the serum antibody responses after mRNA vaccine boosting of mice and human clinical trial participants. In mice, a single dose of a preclinical version of mRNA-1273 vaccine encoding Wuhan-1 spike protein minimally imprinted serum responses elicited by Omicron boosters, enabling generation of type-specific antibodies. However, imprinting was observed in mice receiving an Omicron booster after two priming doses of mRNA-1273, an effect that was mitigated by a second booster dose of Omicron vaccine. In both SARS-CoV-2-infected and uninfected humans who received two Omicron-matched boosters after two or more doses of the prototype mRNA-1273 vaccine, spike-binding and neutralizing serum antibodies cross-reacted with Omicron variants as well as more distantly related sarbecoviruses. Because serum neutralizing responses against Omicron strains and other sarbecoviruses were abrogated after pre-clearing with Wuhan-1 spike protein, antibodies induced by XBB.1.5 boosting in humans focus on conserved epitopes targeted by the antecedent mRNA-1273 primary series. Thus, the antibody response to Omicron-based boosters in humans is imprinted by immunizations with historical mRNA-1273 vaccines, but this outcome may be beneficial as it drives expansion of cross-neutralizing antibodies that inhibit infection of emerging SARS-CoV-2 variants and distantly related sarbecoviruses.
Collapse
Affiliation(s)
- Chieh-Yu Liang
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Saravanan Raju
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Zhuoming Liu
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Yuhao Li
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | | | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Seth J Zost
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cory M Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shayne F Andrew
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Kathryn E Foulds
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sean P J Whelan
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | | | | | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
20
|
Guan D, Dave S, Ebrahim M, Laroche JA. Factors associated with childhood non-vaccination against COVID-19 in Canada: A national survey analysis. Vaccine X 2024; 18:100478. [PMID: 38572339 PMCID: PMC10988031 DOI: 10.1016/j.jvacx.2024.100478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2024] Open
Abstract
Background COVID-19 vaccination efforts are critical in mitigating the impact of the virus, but despite proven safety and efficacy, vaccination rates among children in Canada are lower than in adults, prompting a need to explore determinants of childhood COVID-19 non-vaccination to improve uptake. Method This study analyzed data from the Canadian COVID-19 Immunization Coverage Survey 2022. Using multivariable logistic regression, it examined the association between COVID-19 non-vaccination among children aged 5-17 and factors such as parental sociodemographic characteristics, vaccine knowledge, attitudes, and beliefs (KAB), and vaccination history. Results The analysis revealed that negative KAB towards vaccines, reflected in higher KAB composite scores, significantly increased the likelihood of non-vaccination. Additionally, factors such as lower household incomes, rural residence, employment in sectors not at risk for vaccine-preventable diseases, and younger parental age were associated with higher non-vaccination. The study also highlighted ethnic disparities in vaccination odds and found that children with incomplete routine vaccinations or inconsistent flu vaccination histories were more likely to be unvaccinated against COVID-19. Surprisingly, children of parents who consistently received flu vaccinations were more likely to be unvaccinated against COVID-19. Furthermore, parental education levels showed a complex relationship with children's COVID-19 vaccination status, indicating nuanced influences on vaccination decisions. Conclusion The findings offer vital insights into the factors influencing COVID-19 vaccination uptake among children in Canada, suggesting avenues for targeted strategies to improve vaccine coverage.
Collapse
Affiliation(s)
- David Guan
- Centre for Immunization Surveillance, Public Health Agency of Canada, Ottawa, Ontario, Canada
| | - Sailly Dave
- Centre for Immunization Surveillance, Public Health Agency of Canada, Ottawa, Ontario, Canada
| | - Marwa Ebrahim
- Centre for Immunization Surveillance, Public Health Agency of Canada, Ottawa, Ontario, Canada
| | - Julie A. Laroche
- Centre for Immunization Surveillance, Public Health Agency of Canada, Ottawa, Ontario, Canada
| |
Collapse
|
21
|
Whiting KA, Guest R, Seshan VE, Kamboj M. Susceptibility of healthcare personnel with severe acute respiratory coronavirus virus 2 (SARS-CoV-2) hybrid immunity to XBB lineage reinfection. Infect Control Hosp Epidemiol 2024; 45:781-784. [PMID: 38374682 DOI: 10.1017/ice.2023.282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Among 8,678 vaccinated healthcare personnel (HCP) with previous coronavirus disease 2019 (COVID-19), by August 28, 2023, 909 (10%) had an infection of severe acute respiratory coronavirus virus 2 (SARS-CoV-2) omicron XBB variant. Reinfection risk was comparable irrespective of previous infection type except for the omicron BQ.1 variant. Bivalent vaccination had a protective effect. COVID-19 vaccines remain vital to protect HCP, including those with hybrid immunity.
Collapse
Affiliation(s)
- Karissa A Whiting
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rebecca Guest
- Employee Health, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Venkatraman E Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mini Kamboj
- Infectious Diseases, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Infection Control, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Joan and Sanford Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
22
|
Horváth JK, Túri G, Krisztalovics K, Kristóf K, Oroszi B. Vaccine Effectiveness against GP-Attended Symptomatic COVID-19 and Hybrid Immunity among Adults in Hungary during the 2022-2023 Respiratory Season Dominated by Different SARS-CoV-2 Omicron Subvariants. Vaccines (Basel) 2024; 12:496. [PMID: 38793747 PMCID: PMC11125656 DOI: 10.3390/vaccines12050496] [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: 03/11/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Hungary provides the opportunity to evaluate the effectiveness of COVID-19 vaccination in a setting where naturally acquired immunity and hybrid immunity are likely to play a greater role due to suboptimal vaccination coverage. METHODS A test-negative study was conducted during the 2022-2023 respiratory season at the primary care level to determine the effectiveness of at least one COVID-19 booster dose in preventing medically attended symptomatic RT-PCR-confirmed SARS-CoV-2 infection in adults. Unvaccinated patients were used as a reference group. RESULTS A total of 247 cases and 1073 controls were included in the analysis. CVE was 56.8% (95% CI: 11.9-78.8%) in the population aged 60 years and older and 2.3% (95% CI: -50.0-36.3%) in the younger adults against COVID-19 caused by Omicron subvariants, mainly BA.5, BQ.1, and XBB.1. Self-reported COVID-19 in the 60-365 days prior to the current illness did not confer protection against reinfection without vaccination, but together with booster vaccination, it reduced the risk of COVID-19 by 63.0% (95% CI: -28.0-89.3%) and 87.6% (95% CI: 26.4-97.9%) among the 18-59 and 60+ age groups, respectively. CONCLUSIONS CVE against COVID-19 was moderately high in the 60+ age groups. Because of the benefit of hybrid immunity, persons with previous SARS-CoV-2 infection should still be considered for vaccination campaigns.
Collapse
Affiliation(s)
- Judit Krisztina Horváth
- National Laboratory for Health Security, Epidemiology and Surveillance Centre, Semmelweis University, 1085 Budapest, Hungary; (J.K.H.); (G.T.); (K.K.)
| | - Gergő Túri
- National Laboratory for Health Security, Epidemiology and Surveillance Centre, Semmelweis University, 1085 Budapest, Hungary; (J.K.H.); (G.T.); (K.K.)
| | - Katalin Krisztalovics
- National Laboratory for Health Security, Epidemiology and Surveillance Centre, Semmelweis University, 1085 Budapest, Hungary; (J.K.H.); (G.T.); (K.K.)
| | - Katalin Kristóf
- Institute of Laboratory Medicine, Clinical Microbiology Laboratory, Semmelweis University, 1085 Budapest, Hungary;
| | - Beatrix Oroszi
- National Laboratory for Health Security, Epidemiology and Surveillance Centre, Semmelweis University, 1085 Budapest, Hungary; (J.K.H.); (G.T.); (K.K.)
| |
Collapse
|
23
|
Johnston TS, Li SH, Painter MM, Atkinson RK, Douek NR, Reeg DB, Douek DC, Wherry EJ, Hensley SE. Immunological imprinting shapes the specificity of human antibody responses against SARS-CoV-2 variants. Immunity 2024; 57:912-925.e4. [PMID: 38490198 DOI: 10.1016/j.immuni.2024.02.017] [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: 01/08/2024] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/17/2024]
Abstract
The spike glycoprotein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) continues to accumulate substitutions, leading to breakthrough infections of vaccinated individuals. It remains unclear if exposures to antigenically distant SARS-CoV-2 variants can overcome memory B cell biases established by initial SARS-CoV-2 encounters. We determined the specificity and functionality of antibody and B cell responses following exposure to BA.5 and XBB variants in individuals who received ancestral SARS-CoV-2 mRNA vaccines. BA.5 exposures elicited antibody responses that targeted epitopes conserved between the BA.5 and ancestral spike. XBB exposures also elicited antibody responses that primarily targeted epitopes conserved between the XBB.1.5 and ancestral spike. However, unlike BA.5, a single XBB exposure elicited low frequencies of XBB.1.5-specific antibodies and B cells in some individuals. Pre-existing cross-reactive B cells and antibodies were correlated with stronger overall responses to XBB but weaker XBB-specific responses, suggesting that baseline immunity influences the activation of variant-specific SARS-CoV-2 responses.
Collapse
Affiliation(s)
- Timothy S Johnston
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Shuk Hang Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark M Painter
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Reilly K Atkinson
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Naomi R Douek
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David B Reeg
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | | | - E John Wherry
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Scott E Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Jacobson MA, Blanc PD, Tulsky J, Tilly M, Meister R, Huen W, McNicholas JE. Risk of subsequent SARS-CoV-2 infection among vaccinated employees with or without hybrid immunity acquired early in the Omicron-predominant era of the COVID-19 pandemic. Am J Ind Med 2024; 67:334-340. [PMID: 38316635 DOI: 10.1002/ajim.23570] [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: 06/08/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Hybrid immunity, from COVID-19 vaccination followed by SARS-CoV-2 infection acquired after its Omicron variant began predominating, has provided greater protection than vaccination alone against subsequent infection over 1-3 months of observation. Its longer-term protection is unknown. METHODS We conducted a retrospective cohort study of COVID-19 case incidence among healthcare personnel (HCP) mandated to be vaccinated and report on COVID-19-associated symptoms, high-risk exposures, or known-positive test results to an employee health hotline. We compared cases with hybrid immunity, defined as incident COVID-19 during the first 6 weeks of Omicron-variant predominance (run-in period), to those with immunity from vaccination alone during the run-in period. Time until COVID-19 infection over 13 subsequent months (observation period) was analyzed by standard survival analysis. RESULTS Of 5867 employees, 641 (10.9%, 95% confidence interval [CI]: 10.1%-11.8%) acquired hybrid immunity during the run-in period. Of these, 104 (16.2%, 95% CI: 13.5%-19.3%) experienced new SARS-CoV-2 infection during the 13-month observation period, compared to 2177 (41.7%, 95% CI: 40.3%-43.0%) of the 5226 HCP without hybrid immunity. Time until incident infection was shorter among the latter (hazard ratio: 3.09, 95% CI: 2.54-3.78). CONCLUSIONS In a cohort of vaccinated employees, Omicron-era acquired SARS-CoV-2 hybrid immunity was associated with significantly lower risk of subsequent infection over more than a year of observation-a time period far longer than previously reported and during which three, progressively more resistant, Omicron subvariants became predominant. These findings can inform institutional policy and planning for future COVID-19 additional vaccine dosing requirements for employees, for surveillance programs, and for risk modification efforts.
Collapse
Affiliation(s)
- Mark A Jacobson
- Department of Medicine, Division of Occupational, Environmental, and Climate Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Paul D Blanc
- Department of Medicine, Division of Occupational, Environmental, and Climate Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Jacqueline Tulsky
- Department of Medicine, Division of Occupational, Environmental, and Climate Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Monica Tilly
- Department of Medicine, Division of Occupational, Environmental, and Climate Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Raymond Meister
- Department of Medicine, Division of Occupational, Environmental, and Climate Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Will Huen
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - James E McNicholas
- Department of Medicine, Division of Occupational, Environmental, and Climate Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, California, USA
| |
Collapse
|
25
|
Shen H, Chen D, Li C, Huang T, Ma W. A mini review of reinfection with the SARS-CoV-2 Omicron variant. Health Sci Rep 2024; 7:e2016. [PMID: 38605725 PMCID: PMC11007061 DOI: 10.1002/hsr2.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 02/28/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
Background COVID-19 has caused severe morbidity and mortality worldwide. After the end of the dynamic zero-COVID policy in China in December, 2022, concerns regarding reinfection were raised while little was known due to the lack of surveillance data in this country. Aims This study reviews the probability, risk factors, and severity of severe acute respiratory syndrome coronavirus 2 Omicron variant reinfection, as well as the interval between infections, risk of onward transmission by reinfected cases, and the role of booster vaccination against reinfection. Sources References for this review were identified through searches of PubMed and Web of Science up to September 24, 2023. Results The rate of reinfection ranges from 3.1% to 13.0%. Factors associated with a higher risk of reinfection include being female, having comorbidities, and being unvaccinated. Reinfection with the BA.4 or BA.5 variant occurs approximately 180 days after the initial infection. Reinfections are less clinically severe than primary infections, and there is evidence of lower transmissibility. The debate surrounding the effectiveness and feasibility of booster vaccinations in preventing reinfection continues. Conclusions The reinfection rate during the Omicron epidemic is significantly higher than in previous epidemic periods. However, the symptoms and infectivity of reinfection were weaker than those of the prior infection. Medical staff and individuals at high risk of reinfection should be vigilant. The efficacy of booster vaccinations in reducing reinfection is currently under debate.
Collapse
Affiliation(s)
- Hongwei Shen
- Shenzhen Hospital of Southern Medical UniversityShenzhenGuangdongChina
| | - Dingqiang Chen
- Zhujiang Hospital of Southern Medical UniversityGuangzhouGuangdongChina
| | - Chenglin Li
- Shenzhen Hospital of Southern Medical UniversityShenzhenGuangdongChina
| | - Tingting Huang
- Shenzhen Hospital of Southern Medical UniversityShenzhenGuangdongChina
| | - Wen Ma
- Shenzhen Hospital of Southern Medical UniversityShenzhenGuangdongChina
| |
Collapse
|
26
|
Arantes I, Gomes M, Ito K, Sarafim S, Gräf T, Miyajima F, Khouri R, de Carvalho FC, de Almeida WAF, Siqueira MM, Resende PC, Naveca FG, Bello G. Spatiotemporal dynamics and epidemiological impact of SARS-CoV-2 XBB lineage dissemination in Brazil in 2023. Microbiol Spectr 2024; 12:e0383123. [PMID: 38315011 PMCID: PMC10913747 DOI: 10.1128/spectrum.03831-23] [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: 11/06/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024] Open
Abstract
The SARS-CoV-2 XBB is a group of highly immune-evasive lineages of the Omicron variant of concern that emerged by recombining BA.2-descendent lineages and spread worldwide during 2023. In this study, we combine SARS-CoV-2 genomic data (n = 11,065 sequences) with epidemiological data of severe acute respiratory infection (SARI) cases collected in Brazil between October 2022 and July 2023 to reconstruct the space-time dynamics and epidemiologic impact of XBB dissemination in the country. Our analyses revealed that the introduction and local emergence of lineages carrying convergent mutations within the Spike protein, especially F486P, F456L, and L455F, propelled the spread of XBB* lineages in Brazil. The average relative instantaneous reproduction numbers of XBB* + F486P, XBB* + F486P + F456L, and XBB* + F486P + F456L + L455F lineages in Brazil were estimated to be 1.24, 1.33, and 1.48 higher than that of other co-circulating lineages (mainly BQ.1*/BE*), respectively. Despite such a growth advantage, the dissemination of these XBB* lineages had a reduced impact on Brazil's epidemiological scenario concerning previous Omicron subvariants. The peak number of SARI cases from SARS-CoV-2 during the XBB wave was approximately 90%, 80%, and 70% lower than that observed during the previous BA.1*, BA.5*, and BQ.1* waves, respectively. These findings revealed the emergence of multiple XBB lineages with progressively increasing growth advantage, yet with relatively limited epidemiological impact in Brazil throughout 2023. The XBB* + F486P + F456L + L455F lineages stand out for their heightened transmissibility, warranting close monitoring in the months ahead. IMPORTANCE Brazil was one the most affected countries by the SARS-CoV-2 pandemic, with more than 700,000 deaths by mid-2023. This study reconstructs the dissemination of the virus in the country in the first half of 2023, a period characterized by the dissemination of descendants of XBB.1, a recombinant of Omicron BA.2 lineages evolved in late 2022. The analysis supports that XBB dissemination was marked by the continuous emergence of indigenous lineages bearing similar mutations in key sites of their Spike protein, a process followed by continuous increments in transmissibility, and without repercussions in the incidence of severe cases. Thus, the results suggest that the epidemiological impact of the spread of a SARS-CoV-2 variant is influenced by an intricate interplay of factors that extend beyond the virus's transmissibility alone. The study also underlines the need for SARS-CoV-2 genomic surveillance that allows the monitoring of its ever-shifting composition.
Collapse
Affiliation(s)
- Ighor Arantes
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Marcelo Gomes
- Grupo de Métodos Analíticos em Vigilância Epidemiológica, Fiocruz, Rio de Janeiro, Brazil
| | - Kimihito Ito
- International Institute for Zoonosis Control, Hokkaido University, Hokkaido, Japan
| | - Sharbilla Sarafim
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Tiago Gräf
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fiocruz, Curitiba, 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
| | - 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
| | - 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
| | - Gonzalo Bello
- Laboratório de Arbovírus e Vírus Hemorrágicos, 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
- Grupo de Métodos Analíticos em Vigilância Epidemiológica, Fiocruz, Rio de Janeiro, Brazil
- International Institute for Zoonosis Control, Hokkaido University, Hokkaido, Japan
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fiocruz, Curitiba, Brazil
- Fiocruz, Fortaleza, Brazil
- Instituto Gonçalo Moniz, Fiocruz, Salvador, Brazil
- Departamento do Programa Nacional de Imunizações, Coordenação-Geral de Vigilância das doenças imunopreveníveis, Secretaria de Vigilância em saúde e ambiente, Brasília, Brazil
- Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, 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
| |
Collapse
|
27
|
Kahn F, Bonander C, Moghaddassi M, Christiansen CB, Bennet L, Malmqvist U, Inghammar M, Björk J. Previous SARS-CoV-2 infections and their impact on the protection from reinfection during the Omicron BA.5 wave - a nested case-control study among vaccinated adults in Sweden. IJID REGIONS 2024; 10:235-239. [PMID: 38532742 PMCID: PMC10964055 DOI: 10.1016/j.ijregi.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/28/2024]
Abstract
Objectives We evaluated the protection afforded by SARS-CoV-2 infection-induced immunity against reinfection among working-age vaccinated individuals during a calendar period from June to December 2022 when Omicron BA.5 was the dominating subvariant in Scania County, Sweden. Methods The study cohort (n = 71,592) mainly consisted of health care workers. We analyzed 4144 infected cases during the Omicron BA.5 dominance and 41,440 sex- and age-matched controls with conditional logistic regression. Results The average protection against reinfection was marginal (16%, 95% confidence interval [CI] 7-23%) during the study period but substantially higher for recent infections. Recent infection (3-6 months) with Omicron BA.2 and BA.5 offered strong protection (86%, 95% CI 68-94% and 78%, 95% CI 69-84%), whereas more distant infection (6-12 months) with Omicron BA.1, BA.2, and the variants before Omicron offered marginal or no protection. Conclusions These findings suggest that infection-induced immunity contributes to short-term population protection against infection with the subvariant BA.5 among working-age vaccinated individuals but wanes considerably with time, independent of the virus variant.
Collapse
Affiliation(s)
- Fredrik Kahn
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Carl Bonander
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Mahnaz Moghaddassi
- Department of Clinical Sciences Malmö, Section for Social Medicine and Global Health, Lund University, Malmö, Sweden
| | - Claus Bohn Christiansen
- Department of Clinical Microbiology and Infection Prevention and Control, Skåne University Hospital, Lund, Sweden
| | - Louise Bennet
- Department of Clinical Sciences Malmö, Section for Family Medicine, Lund University, Malmö, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Ulf Malmqvist
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Malin Inghammar
- Department of Clinical Sciences Lund, Section for Infection Medicine, Lund University, Lund, Sweden
| | - Jonas Björk
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Aguilar Ticona JP, Xiao M, Li D, Nery N, Hitchings M, Belitardo EMMA, Fofana MO, Victoriano R, Cruz JS, de Moraes L, Strobel IM, Silva JJ, Sena do Aragão Filho A, Ribeiro GS, Reis MG, Costa F, Khouri R, Ko AI, Cummings DAT. Extensive transmission of SARS-CoV-2 BQ.1* variant in a population with high levels of hybrid immunity: A prevalence survey. Int J Infect Dis 2024; 139:159-167. [PMID: 38070701 PMCID: PMC10784150 DOI: 10.1016/j.ijid.2023.11.039] [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: 07/25/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/01/2024] Open
Abstract
OBJECTIVES The SARS-CoV-2 BQ.1* variant rapidly spread globally in late 2022, posing a challenge due to its increased immune evasion. METHODS We conducted a prevalence survey in Brazil from November 16 to December 22, 2022, as part of a cohort study. We conducted interviews and collected nasal samples for reverse transcription-polymerase chain reaction (RT-PCR) testing and whole-genome sequencing. Cumulative incidence was estimated using RT-PCR positivity, cycle threshold values, and external data on the dynamics of RT-PCR positivity following infection. RESULTS Among 535 participants, 54% had documented SARS-CoV-2 exposure before this outbreak and 74% had received COVID-19 vaccination. In this study, 14.8% tested positive for SARS-CoV-2, with BQ.1* identified in 90.7% of cases. Using case data and cycle threshold values, cumulative incidence was estimated at 56% (95% confidence interval, 36-88%). Of the 79 positive participants, 48.1% had a symptomatic illness, with a lower proportion fulfilling the World Health Organization COVID-19 case definition compared to prior Omicron waves. No participants required medical attention. CONCLUSIONS Despite high population-level hybrid immunity, the BQ.1* variant attacked 56% of our population. Lower disease severity was associated with BQ.1* compared to prior Omicron variants. Hybrid immunity may provide protection against future SARS-CoV-2 variants but in this case was not able to prevent widespread transmission.
Collapse
Affiliation(s)
- Juan P Aguilar Ticona
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil; Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States.
| | - Meng Xiao
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States; Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Li
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States; Public Health Emergency Center, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Nivison Nery
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil; Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States
| | - Matt Hitchings
- Department of Biostatistics, University of Florida, Gainesville, United States; Emerging Pathogens Institute, University of Florida, Gainesville, United States
| | | | - Mariam O Fofana
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States
| | - Renato Victoriano
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil
| | - Jaqueline S Cruz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil
| | - Laise de Moraes
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil
| | - Icaro Morais Strobel
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil
| | - Jessica Jesus Silva
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil
| | | | - Guilherme S Ribeiro
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil; Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Mitermayer G Reis
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States; Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
| | - Federico Costa
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil; Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States
| | - Ricardo Khouri
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil
| | - Albert I Ko
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Salvador, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, United States
| | - Derek A T Cummings
- Department of Biology, University of Florida, Gainesville, United States; Emerging Pathogens Institute, University of Florida, Gainesville, United States
| |
Collapse
|
29
|
Röltgen K, Boyd SD. Antibody and B Cell Responses to SARS-CoV-2 Infection and Vaccination: The End of the Beginning. ANNUAL REVIEW OF PATHOLOGY 2024; 19:69-97. [PMID: 37738512 DOI: 10.1146/annurev-pathmechdis-031521-042754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
As the COVID-19 pandemic has evolved during the past years, interactions between human immune systems, rapidly mutating and selected SARS-CoV-2 viral variants, and effective vaccines have complicated the landscape of individual immunological histories. Here, we review some key findings for antibody and B cell-mediated immunity, including responses to the highly mutated omicron variants; immunological imprinting and other impacts of successive viral antigenic variant exposures on antibody and B cell memory; responses in secondary lymphoid and mucosal tissues and non-neutralizing antibody-mediated immunity; responses in populations vulnerable to severe disease such as those with cancer, immunodeficiencies, and other comorbidities, as well as populations showing apparent resistance to severe disease such as many African populations; and evidence of antibody involvement in postacute sequelae of infection or long COVID. Despite the initial phase of the pandemic ending, human populations will continue to face challenges presented by this unpredictable virus.
Collapse
Affiliation(s)
- Katharina Röltgen
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA;
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
30
|
Johnston TS, Li SH, Painter MM, Atkinson RK, Douek NR, Reeg DB, Douek DC, Wherry EJ, Hensley SE. Immunological imprinting shapes the specificity of human antibody responses against SARS-CoV-2 variants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.08.24301002. [PMID: 38260304 PMCID: PMC10802657 DOI: 10.1101/2024.01.08.24301002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The spike glycoprotein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) continues to accumulate substitutions, leading to breakthrough infections of vaccinated individuals and prompting the development of updated booster vaccines. Here, we determined the specificity and functionality of antibody and B cell responses following exposure to BA.5 and XBB variants in individuals who received ancestral SARS-CoV-2 mRNA vaccines. BA.5 exposures elicited antibody responses that primarily targeted epitopes conserved between the BA.5 and ancestral spike, with poor reactivity to the XBB.1.5 variant. XBB exposures also elicited antibody responses that targeted epitopes conserved between the XBB.1.5 and ancestral spike. However, unlike BA.5, a single XBB exposure elicited low levels of XBB.1.5-specific antibodies and B cells in some individuals. Pre-existing cross-reactive B cells and antibodies were correlated with stronger overall responses to XBB but weaker XBB-specific responses, suggesting that baseline immunity influences the activation of variant-specific SARS-CoV-2 responses.
Collapse
Affiliation(s)
- Timothy S. Johnston
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- Vaccine Research Center, NIAID, NIH; Bethesda, MD
- These authors contributed equally
| | - Shuk Hang Li
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- These authors contributed equally
| | - Mark M. Painter
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- These authors contributed equally
| | - Reilly K. Atkinson
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
| | - Naomi R. Douek
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
| | - David B. Reeg
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | | | - E. John Wherry
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
| | - Scott E. Hensley
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA
| |
Collapse
|
31
|
Maeda H, Saito N, Igarashi A, Ishida M, Terada M, Masuda S, Osawa R, Hosokawa N, Nakashima K, Kamura H, Imura H, Inoue H, Matsuzaka S, Sugimoto Y, Kuwamitsu O, Motohashi I, Morikawa T, Oda R, Hoshina Y, Matono T, Teshigahara O, Sando E, Asami S, Kudo S, Akizuki N, Muto Y, Hayakawa T, Kishaba T, Ohara Y, Kubo Y, Suzuki M, Morimoto K. Effectiveness of primary series, first, and second booster vaccination of monovalent mRNA COVID-19 vaccines against symptomatic SARS-CoV-2 infections and severe diseases during the SARS-CoV-2 omicron BA.5 epidemic in Japan: vaccine effectiveness real-time surveillance for SARS-CoV-2 (VERSUS). Expert Rev Vaccines 2024; 23:213-225. [PMID: 38288980 DOI: 10.1080/14760584.2024.2310807] [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: 12/11/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND This study aimed to evaluate VE of primary, first, and second booster ancestral-strain monovalent mRNA COVID-19 vaccination against symptomatic infections and severe diseases in Japan. METHODS We conducted a test-negative case-control study. We included medically attended episodes and hospitalizations involving individuals aged ≥ 16 with signs and symptoms from July to November 2022, when Omicron BA.5 was dominant nationwide. To evaluate VE, we calculated adjusted ORs of vaccination among test-positive versus test-negative individuals using a mixed-effects logistic regression. RESULTS For VE against symptomatic infections among individuals aged 16 to 59, VE of primary vaccination at > 180 days was 26.1% (95% CI: 10.6-38.8%); VE of the first booster was 58.5% (48.4-66.7%) at ≤ 90 days, decreasing to 41.1% (29.5-50.8%) at 91 to 180 days. For individuals aged ≥ 60, VE of the first booster was 42.8% (1.7-66.7%) at ≤ 90 days, dropping to 15.4% (-25.9-43.2%) at 91 to 180 days, and then increasing to 44.0% (16.4-62.5%) after the second booster. For VE against severe diseases, VE of the first and second booster was 77.3% (61.2-86.7%) at ≤ 90 days and 55.9% (23.4-74.6%) afterward. CONCLUSION mRNA booster vaccination provided moderate protection against symptomatic infections and high-level protection against severe diseases during the BA.5 epidemic in Japan.
Collapse
Affiliation(s)
- Haruka Maeda
- Department of Respiratory Infections, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Department of Clinical Tropical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Nobuo Saito
- Kenya Research Station, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Ataru Igarashi
- School of Data Science, Yokohama City University School of Medicine, Kanagawa, Japan
- Department of Health Economics and Outcomes Research, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Masayuki Ishida
- Department of Infectious Disease Medicine, Chikamori Hospital, Kochi, Japan
| | - Mayumi Terada
- Department of Internal Medicine, Nijigaoka Hospital, Nagasaki, Japan
| | - Shingo Masuda
- Department of Infectious Diseases, Nagasaki University Hospital, Nagasaki, Japan
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Ryosuke Osawa
- Department of Infectious Diseases, Kameda Medical Center, Chiba, Japan
| | - Naoto Hosokawa
- Department of Infectious Diseases, Kameda Medical Center, Chiba, Japan
| | - Kei Nakashima
- Department of Pulmonology, Kameda Medical Center, Chiba, Japan
| | | | - Haruki Imura
- Department of Infectious Disease Medicine, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Hiroki Inoue
- Department of Infectious Disease Medicine, Rakuwakai Otowa Hospital, Kyoto, Japan
| | - Suguru Matsuzaka
- Department of General Medicine, Fukuoka Seishukai Hospital, Fukuoka, Japan
| | - Yukihiro Sugimoto
- Department of Respiratory Medicine, Fukuoka Seishukai Hospital, Fukuoka, Japan
| | | | - Iori Motohashi
- Department of General Internal Medicine, Kawasaki Municipal Tama Hospital, Kawasaki, Japan
| | - Toru Morikawa
- Department of General Medicine, Nara City Hospital, Nara, Japan
| | - Rentaro Oda
- Department of Infectious Diseases, Tokyo Bay Urayasu Ichikawa Medical Center, Chiba, Japan
| | - Yuiko Hoshina
- Department of Infectious Diseases, Tokyo Bay Urayasu Ichikawa Medical Center, Chiba, Japan
| | - Takashi Matono
- Department of Infectious Diseases, Aso Iizuka Hospital, Fukuoka, Japan
- Division of Infectious Disease and Hospital Epidemiology, Saga University Hospital, Saga, Japan
| | | | - Eiichiro Sando
- Department of General Internal Medicine and Clinical Infectious Diseases, Kita-Fukushima Medical Center, Date, Japan
- Department of General Internal Medicine and Clinical Infectious Diseases, Fukushima Medical University, Fukushima, Japan
| | - Sadaharu Asami
- Department of Cardiology, Musashino Tokushukai Hospital, Tokyo, Japan
| | - Satoshi Kudo
- Department of Nursing, Musashino Tokushukai Hospital, Tokyo, Japan
| | - Noboru Akizuki
- Department of Emergency Medicine, Musashino Tokushukai Hospital, Tokyo, Japan
| | - Yoshikazu Muto
- Department of Infectious Diseases, Tosei General Hospital, Seto, Japan
| | | | - Tomoo Kishaba
- Department of Respiratory Medicine, Okinawa Chubu Hospital, Okinawa, Japan
| | | | - Yoshinao Kubo
- Department of Clinical Tropical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Motoi Suzuki
- Infectious Disease Surveillance Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Konosuke Morimoto
- Department of Respiratory Infections, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
32
|
Li L, Xie Z, Li Y, Luo M, Zhang L, Feng C, Tang G, Huang H, Hou R, Xu Y, Jia S, Shi J, Fan Q, Gan Q, Yu N, Hu F, Li Y, Lan Y, Tang X, Li F, Deng X. Immune response and severity of Omicron BA.5 reinfection among individuals previously infected with different SARS-CoV-2 variants. Front Cell Infect Microbiol 2023; 13:1277880. [PMID: 38188634 PMCID: PMC10766752 DOI: 10.3389/fcimb.2023.1277880] [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: 08/15/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction COVID-19 continues to spread worldwide, with an increasing number of individuals experiencing reinfection after recovering from their primary infection. However, the nature and progression of this infection remain poorly understood. We aimed to investigate the immune response, severity and outcomes of Omicron BA.5 reinfection among individuals previously infected with different SARS-CoV-2 variants. Methods We enrolled 432 COVID-19 cases who had experienced prior infection with the ancestral SARS-CoV-2 virus, Delta variant or Omicron BA.2 variant between January 2020 and May 2022 in Guangzhou, China. All cases underwent follow-up from March to April, 2023 through telephone questionnaires and clinical visits. Nasal lavage fluid and peripheral blood were collected to assess anti-RBD IgA, anti-RBD IgG and virus-specific IFN-γ secreting T cells. Results Our study shows that 73.1%, 56.7% and 12.5% of individuals with a prior infection of the ancestral virus, Delta or Omicron BA.2 variant experienced reinfection with the BA.5 variant, respectively. Fever, cough and sore throat were the most common symptoms of BA.5 reinfection, with most improving within one week and none progressing to a critical condition. Compared with individuals without reinfection, reinfected patients with a prior Delta infection exhibited elevated levels of nasal anti-RBD IgA, serum anti-RBD IgG and IFN-γ secreting T cells, whereas there was no noticeable change in reinfected individuals with a prior BA.2 infection. Conclusion These results suggest that BA.5 reinfection is common but severe outcomes are relatively rare. Reinfection with a novel SARS-CoV-2 variant different from the prior infection may induce a more robust immune protection, which should be taken into account during vaccine development.
Collapse
Affiliation(s)
- Lu Li
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiwei Xie
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Youxia Li
- Department of Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Minhan Luo
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lieguang Zhang
- Department of Radiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chengqian Feng
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guofang Tang
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huang Huang
- Department of Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruitian Hou
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yujuan Xu
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shijie Jia
- Department of Traditional Chinese Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingrong Shi
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qinghong Fan
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qingxin Gan
- Department of Radiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Na Yu
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fengyu Hu
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Bio-Island, Guangzhou, China
| | - Yueping Li
- Department of Infectious Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yun Lan
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoping Tang
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Bio-Island, Guangzhou, China
| | - Feng Li
- Institute of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Bio-Island, Guangzhou, China
| | - Xilong Deng
- Department of Critical Care Medicine, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
33
|
Kitamura N, Otani K, Kinoshita R, Yan F, Takizawa Y, Fukushima K, Yoneoka D, Suzuki M, Kamigaki T. Protective effect of previous infection and vaccination against reinfection with BA.5 Omicron subvariant: a nationwide population-based study in Japan. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2023; 41:100911. [PMID: 38223396 PMCID: PMC10786644 DOI: 10.1016/j.lanwpc.2023.100911] [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: 05/29/2023] [Revised: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 01/16/2024]
Abstract
Background The Omicron variant of SARS-CoV-2 was reported to evade immunity derived from vaccination and previous infection. A better understanding of hybrid immunity informs effective infection control strategies. Since the reinfection risk was not well-assessed in East Asia, this study aims to evaluate the risk of infection with Omicron subvariant BA.5 among previously infected individuals in Japan. Methods All notified cases were extracted from the Japanese national COVID-19 surveillance database including 20,297,335 records up to 25 September 2022. Reinfection with BA.5 was defined as the infection notified during the BA.5 dominated period with any prior SARS-CoV-2 infection. The protective effect of prior infections against reinfections with BA.5 was estimated by applying a case-population design and the protective effect of vaccination was estimated by a multivariable Cox regression adjusting for age, sex, variants of prior infection, and the time since the last vaccination. Findings Among 19,830,548 SARS-CoV-2 first infections, 233,424 (1.2%) were reinfected with BA.5. The protective effect against BA.5 reinfection of prior infection with Wuhan strain was 46%, Alpha variant was 35%, Delta variant was 41%, and BA.1/BA.2 subvariant was 74%. The reduced risk of BA.5 reinfection by 7%, 33%, and 66% was associated with two, three, and four doses of vaccination, respectively, compared with one-dose vaccination. Interpretation The prior infections with Omicron subvariant BA.1/BA.2 protected BA.5 reinfection more than pre-Omicron variants. Increased frequency of vaccination led to more protection from reinfection with BA.5. Up-to-date vaccination may be encouraged to prevent future reinfection among the previously infected population. Funding None.
Collapse
Affiliation(s)
- Noriko Kitamura
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kanako Otani
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ryo Kinoshita
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Fangyu Yan
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yu Takizawa
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kohei Fukushima
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Daisuke Yoneoka
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Motoi Suzuki
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Taro Kamigaki
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
34
|
Arbel R, Wolff-Sagy Y. The superiority of bivalent over monovalent booster vaccines. THE LANCET. INFECTIOUS DISEASES 2023; 23:1324-1325. [PMID: 37543043 DOI: 10.1016/s1473-3099(23)00424-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 08/07/2023]
Affiliation(s)
- Ronen Arbel
- Community Medical Services Division, Clalit Health Services, Tel Aviv, Israel; Maximising Health Outcomes Research Lab, Sapir College, Sderot, Israel.
| | - Yael Wolff-Sagy
- Branch of Planning and Strategy, Clalit Health Services, Tel Aviv, Israel
| |
Collapse
|
35
|
Hannawi S, Yan L, Saifeldin L, Abuquta A, Alamadi A, Mahmoud SA, Hassan A, Zhang M, Gao C, Chen Y, Gai W, Xie L. Safety and immunogenicity of multivalent SARS-CoV-2 protein vaccines: a randomized phase 3 trial. EClinicalMedicine 2023; 64:102195. [PMID: 37731938 PMCID: PMC10507195 DOI: 10.1016/j.eclinm.2023.102195] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023] Open
Abstract
Background COVID-19 vaccines that offer broad-spectrum protection are needed. We aimed to evaluate the safety and immunogenicity of multivalent vaccines, SCTV01E and SCTV01C, and compare them with an inactivated vaccine. Methods In the phase 3 trial (ClinicalTrials.gov: NCT05323461), adult participants previously vaccinated with Sinopharm's inactivated SARS-CoV-2 vaccine (BBBIP-CorV) were assigned to receive one booster dose of BBBIP-CorV, 20 μg SCTV01C, or 30 μg SCTV01E. The primary endpoint was to evaluate the geometric mean titers (GMT) of neutralizing antibody (nAb) against the Delta and Omicron BA.1 variants on day 28 after injection. Additional endpoints included GMTs of nAb against Delta (B.1.617.2) and Omicron BA.1 variants on day 180, GMTs against BA.5 on day 28, as well as solicited adverse events (AEs) within seven days, unsolicited AEs within 28 days, and serious AEs, AEs of special interest within 180 days after vaccination. Findings Between May 30, 2022 and October 28, 2022, a total of 1351 participants were randomized to BBBIP-CorV, SCTV01C, or SCTV01E in a 1:1:1 ratio, with immunogenicity assessments performed on the first 300 participants. For BBBIP-CorV, SCTV01C, and SCTV01E groups, the day 28 GMTs of neutralizing antibody against Omicron BA.1 were a 2.38-, 19.37-, and 28.06-fold increase from baseline; the GMTs against Omicron BA.5 were 2.07-, 15.89- and 21.11-fold increases; the GMTs against Delta variants were 1.97-, 12.76-, and 15.88-fold increases, respectively. The day 28 geometric mean ratio (GMR) of SCTV01C/BBIBP-CorV for Omicron BA.1 was 6.49 (95% CI: 4.75, 8.88), while the GMR of SCTV01E/BBIBP-CorV was 9.56 (95% CI: 6.85, 13.33). For the Delta variant, the day 28 GMR of SCTV01C/BBIBP-CorV was 6.26 (95% CI: 4.78, 8.19), and the day 28 GMR of SCTV01E/BBIBP-CorV was 7.26 (95% CI: 5.51, 9.56). On Day 180, the GMTs against Omicron BA.1 were 2.80-, 9.51-, and 15.56-fold increase from baseline, while those against Delta were 1.58-, 5.49-, and 6.63-fold for BBBIP-CorV, SCTV01C, and SCTV01E groups, respectively. Subgroup analyses showed that SCTV01C and SCTV01E induced uniformly high GMTs against both BA.1 and BA.5, demonstrating its superiority over BBIBP-CorV, regardless of baseline GMT levels. Safety and reactogenicity were similar among the three vaccines. Most AEs were Grade 1 or 2. There were 15 ≥Grade 3 AEs: 6 in the BBIBP-CorV group, 4 in the SCTV01C group and 5 in the SCTV01E group. No SAE was reported and one grade 1 AESI (Bell's palsy) was observed in SCTV01C group. Interpretation A booster dose of the tetravalent vaccine SCTV01E consistently induced high neutralizing antibody responses against Omicron BA.1, BA.5, and Delta variants, demonstrating superiority over inactivated vaccine. There is evidence to suggest that SCTV01E may have GMT superiority over bivalent vaccine SCTV01C against Delta, BA.1 and BA.5 variants. Funding This study was sponsored by Sinocelltech Ltd., and funded by the Beijing Science and Technology Planning Project [Z221100007922012] and the National Key Research and Development Program of China [2022YFC0870600].
Collapse
Affiliation(s)
- Suad Hannawi
- Internal Medicine Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai, United Arab Emirates
| | - Lixin Yan
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Linda Saifeldin
- General Surgery Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai, United Arab Emirates
| | - Alaa Abuquta
- Internal Medicine Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai, United Arab Emirates
| | - Ahmad Alamadi
- Ear, Nose and Throat Department (ENT), Al Kuwait-Dubai (ALBaraha) Hospital, Dubai, United Arab Emirates
| | | | - Aala Hassan
- Internal Medicine Department, Al Kuwait-Dubai (ALBaraha) Hospital, Dubai, United Arab Emirates
| | - Miaomiao Zhang
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Cuige Gao
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Yuanxin Chen
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Wenlin Gai
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| | - Liangzhi Xie
- Beijing Engineering Research Center of Protein and Antibody, Sinocelltech Ltd., Beijing, China
| |
Collapse
|
36
|
Al-Dury S, Waldenström J, Ringlander J, Einarsdottir S, Andersson M, Hamah Saed H, Waern J, Martner A, Hellstrand K, Lagging M. Catch-up antibody responses and hybrid immunity in mRNA vaccinated patients at risk of severe COVID-19. Infect Dis (Lond) 2023; 55:744-750. [PMID: 37395287 DOI: 10.1080/23744235.2023.2230289] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND The immunogenicity of repeated vaccination and hybrid immunity in vulnerable patients remains unclear. METHODS We studied the impact of iterative Covid-19 mRNA vaccination and hybrid immunity on antibody levels in immunosuppressed subjects. Patients with liver cirrhosis (n = 38), survivors of allogeneic haematopoietic stem cell transplantation (allo-HSCT) (n = 36) and patients with autoimmune liver disease (n = 14) along with healthy controls (n = 20) were monitored for SARS-CoV-2-S1 IgG after their 1st-3rd vaccine doses, 31 of whom became infected with the Omicron variant after the 2nd dose. Ten uninfected allo-HSCT recipients received an additional 4th vaccine dose. RESULTS Unexpectedly, immunosuppressed patients achieved antibody levels in parity with controls after the 3rd vaccine dose. In all study cohorts, hybrid immunity (effect of vaccination and natural infection) resulted in approximately 10-fold higher antibody levels than vaccine-induced immunity alone. CONCLUSIONS Three doses of the Covid-19 mRNA vaccine entailed high antibody concentrations even in immunocompromised individuals, and hybrid-immunity resulted further augmented levels than vaccination alone. Clinical trial registration: EudraCT 2021-000349-42.
Collapse
Affiliation(s)
- Samer Al-Dury
- Department of Medicine, Gastroenterology and Hepatology Unit, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jesper Waldenström
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Johan Ringlander
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Sigrun Einarsdottir
- Department of Hematology and Coagulation, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Markus Andersson
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hevar Hamah Saed
- Department of Medicine, Gastroenterology and Hepatology Unit, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Waern
- Department of Medicine, Gastroenterology and Hepatology Unit, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Martner
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristoffer Hellstrand
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Martin Lagging
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
37
|
Mimura W, Ishiguro C, Terada-Hirashima J, Matsunaga N, Maeda M, Murata F, Fukuda H. Bivalent Vaccine Effectiveness Among Adults Aged ≥65 Years During the BA.5-Predominant Period in Japan: The VENUS Study. Open Forum Infect Dis 2023; 10:ofad475. [PMID: 37869405 PMCID: PMC10588616 DOI: 10.1093/ofid/ofad475] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Indexed: 10/24/2023] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron BA.5 became prevalent in July 2022 in Japan. Bivalent messenger RNA (mRNA) vaccines were approved as booster doses for individuals who received the primary series or booster dose by monovalent vaccines. We aimed to assess the effectiveness of bivalent vaccines in Japanese adults aged ≥65 years. Methods We conducted a population-based cohort study using data collected from January 2019 to February 2023 in Japan. We included individuals aged ≥65 years in a municipality who received the first or second booster dose of monovalent mRNA vaccines. We estimated the effectiveness of the second or third booster dose of bivalent mRNA vaccines during the Omicron BA.5-predominant period (July-December 2022), compared with ≥90 days after the booster dose of monovalent vaccines. We used a Cox proportional hazard regression model with vaccination status as a time-dependent covariate. Results A total of 81 977 individuals aged ≥65 years (mean [standard deviation] age, 78.3 [7.4] years; 33 487 male [40.8%]) were included in the study cohort. Among them, 57 396 were vaccinated with the second or third dose of bivalent vaccines (BA.1 or BA.4/5). The effectiveness against coronavirus disease 2019 (COVID-19) was estimated to be 57.9% (95% confidence interval, 52.7%-62.5%) for ≥14 days after the second or third bivalent booster dose, compared with 90 days after the first or second monovalent booster dose. Conclusions The study showed that the bivalent mRNA vaccines as the second and third doses would provide protection against COVID-19 among adults ≥65 years in Japan.
Collapse
Affiliation(s)
- Wataru Mimura
- Section of Clinical Epidemiology, Department of Data Science, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Chieko Ishiguro
- Section of Clinical Epidemiology, Department of Data Science, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Junko Terada-Hirashima
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Nobuaki Matsunaga
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Megumi Maeda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Fumiko Murata
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Haruhisa Fukuda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
38
|
Zhu K, Ma S, Chen H, Xie J, Huang D, Ma G, Huang Y. Factors associated with COVID-19 vaccination coverage in hypertensive patients with Omicron infection in Shanghai, China. Hum Vaccin Immunother 2023; 19:2253599. [PMID: 37905728 PMCID: PMC10760500 DOI: 10.1080/21645515.2023.2253599] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/27/2023] [Indexed: 11/02/2023] Open
Abstract
The potential future burden of COVID-19 is determined by the level of susceptibility of the population to infection. The protective effect provided by those previously infected diminishes over several months, while individuals with mixed immunity have the highest degree and persistence of protection. This study aimed to clarify the vaccination status of COVID-19 patients with hypertension and to analyze the characteristics and risk factors of non-vaccinated patients to protect this vulnerable population in the future. The study ultimately enrolled 4576 hypertensive patients with Omicron infection from April 6, 2022, to May 15, 2022. Among them, 3556 patients (77.7%) had received at least one dose of vaccine, and 2058 patients (45.0%) received a booster dose. In the multivariate logistic analysis, male (OR 1.328, 95% CI 1.138-1.550, p < .001), age (60-69 years vs.18-49 years) (OR 0.348, 95% CI 0.270-0.448, p < .001), age (≥70 years vs.18-49 years) (OR 0.130, 95% CI 0.100-0.169, p < .001), diabetes mellitus (OR 0.553, 95% CI 0.463-0.661, p < .001), chronic pulmonary diseases (OR 0.474, 95% CI 0.260-0.863, p = .015), chronic kidney disease (OR 0.177, 95% CI 0.076-0.410, p < .001), and cancer (OR 0.225, 95% CI 0.094-0.535, p = .001) were associated with vaccinated status. The vaccine coverage rate, especially the booster vaccine, was low for hypertensive patients with Omicron infection. Females, increasing age, and coexisting chronic diseases were associated with more inadequate vaccine coverage in hypertensive COVID-19 patients. Targeted interventions are required to address the under-vaccination of diverse hypertensive populations.
Collapse
Affiliation(s)
- Kongbo Zhu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shaolei Ma
- Department of Emergency and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hui Chen
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jianfeng Xie
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Dan Huang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yingzi Huang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
39
|
Fang L, Xu J, Zhao Y, Fan J, Shen J, Liu W, Cao G. The effects of amino acid substitution of spike protein and genomic recombination on the evolution of SARS-CoV-2. Front Microbiol 2023; 14:1228128. [PMID: 37560529 PMCID: PMC10409611 DOI: 10.3389/fmicb.2023.1228128] [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: 05/24/2023] [Accepted: 07/03/2023] [Indexed: 08/11/2023] Open
Abstract
Over three years' pandemic of 2019 novel coronavirus disease (COVID-19), multiple variants and novel subvariants have emerged successively, outcompeted earlier variants and become predominant. The sequential emergence of variants reflects the evolutionary process of mutation-selection-adaption of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Amino acid substitution/insertion/deletion in the spike protein causes altered viral antigenicity, transmissibility, and pathogenicity of SARS-CoV-2. Early in the pandemic, D614G mutation conferred virus with advantages over previous variants and increased transmissibility, and it also laid a conservative background for subsequent substantial mutations. The role of genomic recombination in the evolution of SARS-CoV-2 raised increasing concern with the occurrence of novel recombinants such as Deltacron, XBB.1.5, XBB.1.9.1, and XBB.1.16 in the late phase of pandemic. Co-circulation of different variants and co-infection in immunocompromised patients accelerate the emergence of recombinants. Surveillance for SARS-CoV-2 genomic variations, particularly spike protein mutation and recombination, is essential to identify ongoing changes in the viral genome and antigenic epitopes and thus leads to the development of new vaccine strategies and interventions.
Collapse
Affiliation(s)
- Letian Fang
- Key Laboratory of Biological Defense, Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Medical Bioprotection, Shanghai, China
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Jie Xu
- Department of Foreign Languages, International Exchange Center for Military Medicine, Second Military Medical University, Shanghai, China
| | - Yue Zhao
- Key Laboratory of Biological Defense, Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Medical Bioprotection, Shanghai, China
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Junyan Fan
- Key Laboratory of Biological Defense, Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Medical Bioprotection, Shanghai, China
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Jiaying Shen
- School of Medicine, Tongji University, Shanghai, China
| | - Wenbin Liu
- Key Laboratory of Biological Defense, Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Medical Bioprotection, Shanghai, China
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Guangwen Cao
- Key Laboratory of Biological Defense, Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Medical Bioprotection, Shanghai, China
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| |
Collapse
|
40
|
Shioda K. Tackling covid-19 variants. BMJ 2023; 382:1603. [PMID: 37451814 DOI: 10.1136/bmj.p1603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Affiliation(s)
- Kayoko Shioda
- Department of Global Health, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
41
|
Boyton RJ, Altmann DM. Imprinted hybrid immunity against XBB reinfection. THE LANCET. INFECTIOUS DISEASES 2023; 23:764-765. [PMID: 36924785 PMCID: PMC10010702 DOI: 10.1016/s1473-3099(23)00138-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 03/14/2023]
Affiliation(s)
- Rosemary J Boyton
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK; Lung Division, Royal Brompton Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - Daniel M Altmann
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| |
Collapse
|
42
|
Yung CF, Pang D, Kam KQ, Lye DC, Ong B, Chong CY, Tan KB. BNT162b2 vaccine protection against omicron and effect of previous infection variant and vaccination sequence among children and adolescents in Singapore: a population-based cohort study. THE LANCET. CHILD & ADOLESCENT HEALTH 2023; 7:463-470. [PMID: 37201540 PMCID: PMC10185330 DOI: 10.1016/s2352-4642(23)00101-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Information on variant-specific vaccine protection and the effect of previous infection variant is scarce in children. We aimed to ascertain the level of protection conferred by BNT162b2 COVID-19 vaccination against omicron variant infection (BA.4 or BA.5, and XBB) in a previously infected national paediatric cohort. We also explored the association between sequence of previous infection (variant) and vaccination on protection. METHODS We did a retrospective, population-based cohort study using the national databases of all confirmed SARS-CoV-2 infections, vaccines administered, and demographic records maintained by the Ministry of Health, Singapore. The study cohort consisted of children aged 5-11 years and adolescents aged 12-17 years who had a previous SARS-CoV-2 infection from Jan 1, 2020, to Dec 15, 2022. People who were infected during the pre-delta period or were immunocompromised (received three vaccination doses [children 5-11 years old] and four vaccinations doses [adolescents 12-17 years old]) were excluded. Those who had multiple episodes of infection before the study start date, were not vaccinated before infection but completed three doses, received bivalent mRNA vaccine, or received non-mRNA vaccine doses were also excluded. All SARS-CoV-2 infections confirmed by reverse transcriptase polymerase chain reaction or rapid antigen testing were grouped into delta, BA.1, BA.2, BA.4 or BA.5, or XBB variants using a combination of whole-genome sequencing, S-gene target failure results, and imputation. For BA.4 or BA.5, the study outcome period was June 1-Sept 30, 2022, and for XBB variants the outcome period was Oct 18-Dec 15, 2022. Incidence rate ratios between vaccinated and unvaccinated were derived using adjusted Poisson regressions and vaccine effectiveness was estimated as (1-risk ratio) × 100%. FINDINGS 135 197 people aged 5-17 years (79 332 children and 55 865 adolescents) were included in the cohort for the vaccine effectiveness analysis against omicron BA.4 or BA.5, and 164 704 people aged 5-17 years (97 235 children and 67 469 adolescents) were included for the analysis against omicron XBB. Approximately 47% of participants were female and 53% were male. Among those previously infected, vaccine effectiveness against BA.4 or BA.5 infection in fully vaccinated children (two doses) was 74·0% (95% CI 67·7-79·1) and in adolescents (three doses) was 85·7% (80·2-89·6). Against XBB, protection conferred with full vaccination was lower at 62·8% (95% CI 42·3-76·0) in children and 47·9% (20·2-66·1) in adolescents. In children, receipt of two-dose vaccination before first SARS-CoV-2 infection provided them with the highest protection against subsequent BA.4 or BA.5 infection at 85·3% (95% CI 80·2-89·1); however, this was not shown to be the case for adolescents. First infection variant had an effect on vaccine effectiveness against omicron BA.4 or BA.5 reinfection in the following descending order: BA.2 conferred the highest protection (92·3% [95% CI 88·9-94·7] in children and 96·4% [93·5-98·0] in adolescents) followed by BA.1 (81·9% [75·9-86·4] in children and 95·0% [91·6-97·0] in adolescents), and delta which conferred the lowest protection (51·9% [5·3-75·6] in children and 77·5% [63·9-86·0] in adolescents). INTERPRETATION In previously infected children and adolescents, BNT162b2 vaccination provided additional protection against omicron BA.4 or BA.5 and XBB variants compared with those who remained unvaccinated. Hybrid immunity against XBB was lower than against BA.4 or BA.5, especially in adolescents. Early vaccination of previously uninfected children before their first SARS-CoV-2 exposure could potentially strengthen population immunity resilience against future variants. FUNDING None.
Collapse
Affiliation(s)
- Chee Fu Yung
- Infectious Disease Service, Department of Paediatrics, and SingHealth Duke-NUS Paediatrics Academic Clinical Program, KK Women's and Children's Hospital, Singapore; Duke-NUS Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore.
| | | | - Kai Qian Kam
- Infectious Disease Service, Department of Paediatrics, and SingHealth Duke-NUS Paediatrics Academic Clinical Program, KK Women's and Children's Hospital, Singapore; Duke-NUS Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - David C Lye
- Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore; Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; National Centre for Infectious Diseases, Singapore
| | - Benjamin Ong
- Ministry of Health, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chia Yin Chong
- Infectious Disease Service, Department of Paediatrics, and SingHealth Duke-NUS Paediatrics Academic Clinical Program, KK Women's and Children's Hospital, Singapore; Duke-NUS Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kelvin B Tan
- Duke-NUS Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore; Ministry of Health, Singapore; National Centre for Infectious Diseases, Singapore
| |
Collapse
|
43
|
Carr EJ, Wu MY, Gahir J, Harvey R, Townsley H, Bailey C, Fowler AS, Dowgier G, Hobbs A, Herman L, Ragno M, Miah M, Bawumia P, Smith C, Miranda M, Mears HV, Adams L, Haptipoglu E, O'Reilly N, Warchal S, Sawyer C, Ambrose K, Kelly G, Beale R, Papineni P, Corrah T, Gilson R, Gamblin S, Kassiotis G, Libri V, Williams B, Swanton C, Gandhi S, Lv Bauer D, Wall EC. Neutralising immunity to omicron sublineages BQ.1.1, XBB, and XBB.1.5 in healthy adults is boosted by bivalent BA.1-containing mRNA vaccination and previous Omicron infection. THE LANCET. INFECTIOUS DISEASES 2023; 23:781-784. [PMID: 37290472 PMCID: PMC10241485 DOI: 10.1016/s1473-3099(23)00289-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023]
Affiliation(s)
- Edward J Carr
- The Francis Crick Institute, London, UK; University College London, London, UK
| | - Mary Y Wu
- COVID Surveillance Unit, London, UK; The Francis Crick Institute, London, UK
| | - Joshua Gahir
- The Francis Crick Institute, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre and Clinical Research Facility, London, UK
| | | | - Hermaleigh Townsley
- The Francis Crick Institute, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre and Clinical Research Facility, London, UK
| | - Chris Bailey
- The Francis Crick Institute, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre and Clinical Research Facility, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | - Emine Haptipoglu
- The Francis Crick Institute, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre and Clinical Research Facility, London, UK
| | | | | | | | | | | | - Rupert Beale
- The Francis Crick Institute, London, UK; University College London, London, UK
| | | | - Tumena Corrah
- London Northwest University Healthcare NHS Trust, London, UK
| | - Richard Gilson
- Central and North West London NHS Foundation Trust, London, UK
| | | | - George Kassiotis
- The Francis Crick Institute, London, UK; Department of Infectious Disease, St Mary's Hospital, Imperial College London, London, UK
| | - Vincenzo Libri
- University College London, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre and Clinical Research Facility, London, UK
| | - Bryan Williams
- University College London, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre and Clinical Research Facility, London, UK
| | - Charles Swanton
- The Francis Crick Institute, London, UK; University College London, London, UK
| | - Sonia Gandhi
- The Francis Crick Institute, London, UK; University College London, London, UK
| | | | - Emma C Wall
- The Francis Crick Institute, London, UK; University College London, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre and Clinical Research Facility, London, UK.
| |
Collapse
|
44
|
Ye C, Zhang G, Zhang A, Xin H, Wu K, Li Z, Jia Y, Hao L, Xue C, Wang Y, Xu H, Zhu W, Zhou Y. The Omicron Variant Reinfection Risk among Individuals with a Previous SARS-CoV-2 Infection within One Year in Shanghai, China: A Cross-Sectional Study. Vaccines (Basel) 2023; 11:1146. [PMID: 37514962 PMCID: PMC10386598 DOI: 10.3390/vaccines11071146] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Reinfection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants due to immune escape is challenging for the global response to the pandemic. We estimated the Omicron reinfection prevalence among people who had a previous SARS-CoV-2 infection in Shanghai, China. We conducted a telephone survey in December 2022 with those who had previously been infected with Omicron between March and May 2022. Information on their demographics, coronavirus disease 2019 (COVID-19) testing, and vaccination history was collected. The overall and subgroup reinfection rates were estimated and compared. Among the 1981 respondents who were infected between March and May 2022, 260 had positive nucleic acid or rapid antigen tests in December 2022, with an estimated reinfection rate of 13.1% (95% confidence interval [95% CI]: 11.6-14.6). The reinfection rate for those who had a booster vaccination was 11.4% (95% CI: 9.2-13.7), which was significantly lower than that for those with an incomplete vaccination series (15.2%, 95% CI: 12.3-18.1) (adjusted odds ratio [aOR]: 0.579; 95% CI: 0.412-0.813). Reinfection with the Omicron variant was lower among individuals with a previous SARS-CoV-2 infection and those who had a booster vaccination, suggesting that hybrid immunity may offer protection against reinfection with Omicron sublineages.
Collapse
Affiliation(s)
- Chuchu Ye
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Ge Zhang
- School of Public Health, Dali University, Dali 671003, China
| | - Anran Zhang
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Hualei Xin
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kang Wu
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Zhongjie Li
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100073, China
| | - Yilin Jia
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Lipeng Hao
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Caoyi Xue
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Yuanping Wang
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Hongmei Xu
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Weiping Zhu
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| | - Yixin Zhou
- Shanghai Pudong New Area Center for Disease Control and Prevention, Shanghai 200136, China
| |
Collapse
|
45
|
Goh AXC, Chae SR, Chiew CJ, Tang N, Pang D, Lin C, Tan KB, Lee VJ, Ho ZJM. Characteristics of the omicron XBB subvariant wave in Singapore. Lancet 2023; 401:1261-1262. [PMID: 37061259 DOI: 10.1016/s0140-6736(23)00390-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/31/2023] [Accepted: 02/20/2023] [Indexed: 04/17/2023]
Affiliation(s)
| | - Sae-Rom Chae
- Ministry of Health, Singapore; National Centre for Infectious Diseases, Singapore
| | - Calvin J Chiew
- Ministry of Health, Singapore; National Centre for Infectious Diseases, Singapore
| | | | | | - Cui Lin
- National Centre for Infectious Diseases, Singapore
| | - Kelvin Bryan Tan
- Ministry of Health, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Vernon J Lee
- Ministry of Health, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Zheng Jie Marc Ho
- Ministry of Health, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|