1
|
Yan Y, Halemubieke S, Shan B, Zhao L, Duan Y, Wang Y, Wu M, Bu X, Wang Q, Chang L, Ji H, Sun H, Liu Y, Sun P, Liu Y, Wang L, Li C, Zhang L, Deng X, Wang L. Longitudinal assessment of immunogenicity of inactivated COVID-19 booster immunization and breakthrough infection in blood donors: A multicenter study from 2021 to 2023. Hum Vaccin Immunother 2025; 21:2498828. [PMID: 40323225 PMCID: PMC12054371 DOI: 10.1080/21645515.2025.2498828] [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: 01/13/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Assessing immune responses across diverse populations is essential for refining public health strategies. Blood donors offer valuable insights into community-level immunity. This study aims to investigate immune responses associated with inactivated COVID-19 booster immunization and breakthrough infections in blood donors. This study was conducted in a cohort of blood donors from six centers across five of China's seven major geographical regions, spanning from December 2021 to February 2023. Blood samples were collected before booster vaccination, at 1, 3, and 6 months post-vaccination, as well as 1 month post-infection. SARS-CoV-2-specific antibodies, T cell specific IFNγ levels, and neutralizing antibodies against wild-type and Omicron strains were measured. Platelet count, anti-PF4 antibody, and D-dimer levels were assessed. Demographic characteristics were analyzed to determine their impact on immunogenicity. SARS-CoV-2-specific antibodies and IFNγ levels significantly increased post-booster, peaking one month after immunization. Antibodies continued to decrease at six months, while IFNγ levels remained stable at this point. Pseudovirus neutralization assays revealed elevated neutralizing antibodies following the booster dose, with minimal response to the XBB.1.5 variant. Following Omicron infection, antibody and IFNγ levels surpassed that observed post-booster. Participants aged 36-49 and those over 50 exhibited weaker immune responses post-booster than those ages 18-35, while those with BMI above 28 showed lower IFNγ levels. This study demonstrates the utility of blood donor samples for tracking immunization effectiveness against emerging pathogens, and highlights enhanced immune responses after booster immunization and breakthrough infections, underscoring the need for tailored vaccination strategies for different groups.
Collapse
Affiliation(s)
- Ying Yan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P. R. China
| | - Shana Halemubieke
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P. R. China
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Baifeng Shan
- Department of Blood Screening Laboratory, Taiyuan Blood Center, Taiyuan, Shanxi, P.R. China
| | - Lei Zhao
- Department of Blood Screening Laboratory, Henan Blood Center, Zhengzhou, Henan, P.R. China
| | - Youbin Duan
- Department of Blood Screening Laboratory, Yunnan Blood Center, Kunming, Yunnan, P.R. China
| | - Yifang Wang
- Department of Blood Screening Laboratory, Henan Blood Center, Zhengzhou, Henan, P.R. China
| | - Mingrui Wu
- Department of Blood Quality Management, Sanmenxia Blood Center, Sanmenxia, Henan, P.R. China
| | - Xiaoxiao Bu
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Quaner Wang
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Le Chang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P. R. China
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Huimin Ji
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P. R. China
| | - Huizhen Sun
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P. R. China
| | - Yang Liu
- Department of Blood Screening Laboratory, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, China
| | - Peng Sun
- Department of Blood Screening Laboratory, Dalian Blood Center, Dalian, Liaoning, P.R. China
| | - Ying Liu
- Department of Blood Screening Laboratory, Dalian Blood Center, Dalian, Liaoning, P.R. China
| | - Lin Wang
- Department of Blood Screening Laboratory, Dalian Blood Center, Dalian, Liaoning, P.R. China
| | - Chuanbao Li
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Libo Zhang
- Department of Blood Screening Laboratory, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, China
| | - Xuelian Deng
- Department of Blood Screening Laboratory, Dalian Blood Center, Dalian, Liaoning, P.R. China
| | - Lunan Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, P.R. China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing, P. R. China
- National Center for Clinical Laboratories, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R. China
| |
Collapse
|
2
|
Li H, Liu H, Wu H, Guo C, Zuo W, Zheng Y, Deng X, Xu J, Wang Y, Wang Z, Lu B, Hou B, Cao B. Reading of human acute immune dynamics in omicron SARS-CoV-2 breakthrough infection. Emerg Microbes Infect 2025; 14:2494705. [PMID: 40231451 PMCID: PMC12064115 DOI: 10.1080/22221751.2025.2494705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 04/16/2025]
Abstract
The dynamics of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infections remain unclear, particularly when compared to responses in naive individuals. In this longitudinal prospective cohort study, 13 participants were recruited. Peripheral blood samples were collected every other day until day 7 after symptom onset. Transcriptome sequencing, single-cell sequencing, T-cell receptor (TCR) sequencing, B-cell receptor (BCR) sequencing, Olink proteomics, and antigen-antibody binding experiments were then performed. During the incubation periods of breakthrough infections, peripheral blood exhibited type 2 cytokine response, which shifted to type 1 cytokine response upon symptom onset. Plasma cytokine levels of C-X-C motif chemokine ligand 10, monocyte chemoattractant protein-1, interferon-γ, and interleukin-6 show larger changes in breakthrough infections than naïve infections. The inflammatory response in breakthrough infections rapidly subsided, returning to homeostasis by day 5 after symptom onset. Notably, the levels of monocyte-derived S100A8/A9, previously considered a marker of severe disease, physiologically significantly increased in the early stages of mild cases and persisted until day 7, suggesting a specific biological function. Longitudinal tracking also revealed that antibodies anti-Receptor Binding Domain (anti-RBD) in breakthrough infections significantly increased by day 7 after symptom onset, whereas cytotoxic T lymphocytes appeared by day 5. This study presents a reference for interpreting the immunological response to breakthrough infectious disease in humans.
Collapse
Affiliation(s)
- Haibo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
| | - Hongyu Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Hongping Wu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Chang Guo
- Changping National Laboratory (CPNL), Beijing, People’s Republic of China
| | - Wenting Zuo
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Ying Zheng
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyan Deng
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, People’s Republic of China
| | - Jiuyang Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yeming Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Binghuai Lu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Baidong Hou
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Bin Cao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| |
Collapse
|
3
|
Bryant N, Muehling LM, Wavell K, Teague WG, Woodfolk JA. Rhinovirus as a driver of airway T cell dynamics in children with treatment-refractory recurrent wheeze. JCI Insight 2025; 10:e189480. [PMID: 40337866 DOI: 10.1172/jci.insight.189480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Severe asthma in children is notoriously difficult to treat, and its immunopathogenesis is complex. In particular, the contribution of T cells and relationships to antiviral immunity remain enigmatic. Here, we coupled deep phenotyping with machine learning methods to elucidate the dynamics of T cells in the lower airways of children with treatment-refractory recurrent wheeze, and examine rhinovirus (RV) as a driver. Our strategy revealed a T cell landscape dominated by type 1 and type 17 CD8+ signatures. Interrogation of phenotypic relationships coupled with trajectory mapping identified T cell migratory and differentiation pathways spanning the blood and airways that culminated in tissue residency, and involved transitions between type 1 and type 17 tissue-resident types. These dynamics were reflected in cytokine polyfunctionality. Use of machine learning tools to cross-compare T cell populations that were enriched in the airways of RV-positive children with those induced in the blood following experimental RV challenge precisely pinpointed RV-responsive signatures that contributed to T cell migratory and differentiation pathways. Despite their rarity, these signatures were also detected in the airways of RV-negative children. Together, our results underscore the aberrant nature of type 1 immunity in the airways of children with recurrent wheeze, and implicate an important viral trigger as a driver.
Collapse
Affiliation(s)
- Naomi Bryant
- Department of Medicine
- Department of Microbiology, Immunology, and Cancer Biology, and
| | | | - Kristin Wavell
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - W Gerald Teague
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Judith A Woodfolk
- Department of Medicine
- Department of Microbiology, Immunology, and Cancer Biology, and
| |
Collapse
|
4
|
Wong YC, Hang Ho DH, Zhou R, Zhang R, Woo KF, Cheng WY, Wang T, Du Y, Polly Pang KP, Tai WK, Jin X, Chen Z, Ngai Hung IF. An open-label study on the safety and immunogenicity of a PD-1-enhanced DNA vaccine used as a T cell booster for COVID-19. EBioMedicine 2025; 115:105699. [PMID: 40245494 DOI: 10.1016/j.ebiom.2025.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Inducing T cell responses by vaccines among elderly has been a long-standing challenge. There is a need for effective COVID-19 vaccines to boost waning immunity against emerging SARS-CoV-2 variants, especially for the elderly. This study investigated the safety and immunogenicity of a PD-1-enhanced COVID-19 DNA vaccine (ICCOV™), as a booster vaccine in healthy adults (aged 18-59 years) and elderly (aged 60-75 years). METHODS This open-label, non-randomised Phase 2 study enrolled healthy participants aged 18-75 years who had previously been vaccinated with Sinovac CoronaVac, Pfizer-bioNTech Comirnaty vaccines, or both. Participants were stratified into four cohorts according to age, primary vaccination, and COVID-19 infection history, namely Adult-CoronaVac, Adult-Comirnaty, Adult-Mixed, and Elderly-Mixed cohorts. Participants were administered with a single dose of 2 mg ICCOV intramuscularly followed by electroporation using the proprietary TERESA-EPT-I device. Participants were followed up for 60 days. The primary endpoint was T cell immunogenicity within 28 days post-ICCOV vaccination. The secondary endpoints were safety, T cell and antibody responses within 60 days post-vaccination (ClinicalTrials.govNCT05904054). FINDINGS The study was conducted at Gleneagles Hospital Hong Kong between 30 June and 30 November 2023. In total, 31 participants were enrolled across the Adult-Comirnaty (n = 4), Adult-Mixed (n = 15), and Elderly-Mixed (n = 12) cohorts. All enrolled participants completed the study and were included in safety and immunogenicity analyses. Among these participants, 2 from the Adult-Comirnaty cohort, 9 from the Adult-Mixed cohort, and 4 from the Elderly-Mixed cohort reported a total of 31 adverse events, all in grade 1-2. Pain at the administration site was the most frequently reported (38·7%). The proportion of participants demonstrating an increase of SARS-CoV-2-specific ELISpot T cell responses within 28 days post ICCOV vaccination was 100% (4/4), 80% (12/15), and 75% (9/12) in Adult-Comirnaty, Adult-Mixed, and Elderly-Mixed cohorts, respectively. Single ICCOV vaccination elicited SARS-CoV-2-specific, polyfunctional CD8+ and CD4+ T cells against both ancestral and Omicron strains in all cohorts. The magnitude of responses was not inferior in the elderly, compared to adults. No elevation of antibody responses was detected. INTERPRETATION Single PD-1-enhanced ICCOV booster DNA vaccination did not show major safety concerns. The ICCOV booster elicited cross-reactive T cell responses to multiple SARS-CoV-2 strains, including in the elderly. This report demonstrates the T-cell boosting immunogenicity of ICCOV in the susceptible elderly population. FUNDING Clinical Translational Catalyst, Hong Kong Science & Technology Parks Corporation.
Collapse
Affiliation(s)
- Yik Chun Wong
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Derek Hoi Hang Ho
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Runhong Zhou
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China
| | - Ruiqi Zhang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China
| | - Kin Fai Woo
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Wing Yin Cheng
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Ting Wang
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Yanhua Du
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China
| | - Ka Po Polly Pang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China
| | - Wai Ki Tai
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China
| | - Xia Jin
- Immuno Cure Holding (HK) Limited, Hong Kong Science Park, Hong Kong Special Administrative Region of the People's Republic of China.
| | - Zhiwei Chen
- AIDS Institute, Department of Microbiology and Pandemic Research Alliance Unit, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of the People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region of the People's Republic of China.
| | - Ivan Fan Ngai Hung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China.
| |
Collapse
|
5
|
Brady C, Tipton T, Carnell O, Longet S, Gooch K, Hall Y, Salguero J, Tomic A, Carroll M. A systems biology approach to define SARS-CoV-2 correlates of protection. NPJ Vaccines 2025; 10:69. [PMID: 40229322 PMCID: PMC11997207 DOI: 10.1038/s41541-025-01103-2] [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: 09/20/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
Correlates of protection (CoPs) for SARS-CoV-2 have yet to be sufficiently defined. This study uses the machine learning platform, SIMON, to accurately predict the immunological parameters that reduced clinical pathology or viral load following SARS-CoV-2 challenge in a cohort of 90 non-human primates. We found that anti-SARS-CoV-2 spike antibody and neutralising antibody titres were the best predictors of clinical protection and low viral load in the lung. Since antibodies to SARS-CoV-2 spike showed the greatest association with clinical protection and reduced viral load, we next used SIMON to investigate the immunological features that predict high antibody titres. It was found that a pre-immunisation response to seasonal beta-HCoVs and a high frequency of peripheral intermediate and non-classical monocytes predicted low SARS-CoV-2 spike IgG titres. In contrast, an elevated T cell response as measured by IFNγ ELISpot predicted high IgG titres. Additional predictors of clinical protection and low SARS-CoV-2 burden included a high abundance of peripheral T cells. In contrast, increased numbers of intermediate monocytes predicted clinical pathology and high viral burden in the throat. We also conclude that an immunisation strategy that minimises pathology post-challenge did not necessarily mediate viral control. This would be an important finding to take forward into the development of future vaccines aimed at limiting the transmission of SARS-CoV-2. These results contribute to SARS-CoV-2 CoP definition and shed light on the factors influencing the success of SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Caolann Brady
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom.
| | - Tom Tipton
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
| | - Oliver Carnell
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Stephanie Longet
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- International Center for Infectiology Research (CIRI), Team GIMAP, Claude Bernard Lyon 1 University, Saint-Etienne, France
| | - Karen Gooch
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Yper Hall
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Javier Salguero
- UK Health Security Agency; Porton Down, Salisbury, United Kingdom
| | - Adriana Tomic
- National Emerging Infectious Diseases Laboratories, Boston, MA, USA
- Department of Virology, Immunology & Microbiology, Boston University Medical School, Boston, MA, USA
- Biomedical Engineering, Boston University, College of Engineering, Boston, MA, USA
| | - Miles Carroll
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
6
|
Liu B, Xu L, Wang Y, Hao C, Jiang W. Understanding the unconventional reemergence of M. pneumoniae epidemics during the COVID-19 pandemic. Transl Pediatr 2025; 14:473-479. [PMID: 40225082 PMCID: PMC11982997 DOI: 10.21037/tp-24-482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Since the implementation of coronavirus disease 2019 (COVID-19) restrictions since 2020, the number of Mycoplasma pneumoniae (M. pneumoniae) infections in children has significantly decreased. However, after the end of the COVID pandemic, there has been a notable resurgence in M. pneumoniae infections, which is particularly unusual in terms of both the number of infections and their severity. The purpose of this article is to review the existing evidence and explore theories that underlying the epidemiological shifts of M. pneumoniae following the COVID-19 pandemic, and propose factors contributing to the unconventional resurgence of M. pneumoniae infections. Proposed factors include decline of M. pneumoniae immunity, circulation of different genetic types and emergence of new macrolide-resistant M. pneumoniae (MRMP) variants, immune dysregulation following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and others. Among these factors, the decline in M. pneumoniae immunity and the circulation of different genetic types are considered significant contributors. Further research in bacterial genomics and more robust immunology studies are needed to guide the prevention of M. pneumoniae infections and the allocation of healthcare resources. International cooperation and information sharing are crucial for understanding the epidemiological changes of M. pneumoniae. Further cross-regional collaboration is called to enhance our understanding of the scope of M. pneumoniae outbreaks and facilitate a collective response.
Collapse
Affiliation(s)
- Bingjie Liu
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Lina Xu
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Yuqing Wang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| | - Wujun Jiang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Peluso M, Sandel D, Deitchman A, Kim S, Dalhuisen T, Tummala H, Tibúrcio R, Zemelko L, Borgo G, Singh S, Schwartz K, Deswal M, Williams M, Hoh R, Shimoda M, Narpala S, Serebryannyy L, Khalili M, Vendrame E, SenGupta D, Whitmore LS, Tisoncik-Go J, Gale M, Koup R, Mullins J, Felber B, Pavlakis G, Reeves J, Petropoulos C, Glidden D, Spitzer M, Gama L, Caskey M, Nussenzweig M, Chew K, Henrich T, Yukl S, Cohn L, Deeks S, Rutishauser R. Combination immunotherapy induces post-intervention control of HIV. RESEARCH SQUARE 2025:rs.3.rs-6141479. [PMID: 40166020 PMCID: PMC11957202 DOI: 10.21203/rs.3.rs-6141479/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The identification of therapeutic strategies to induce sustained antiretroviral therapy (ART)-free control of HIV infection is a major priority.1 Combination immunotherapy including HIV vaccination, immune stimulation/latency reversal, and passive transfer of broadly neutralizing antibodies (bNAbs) has shown promise in non-human primate models,2-7 but few studies have translated such approaches into people. Here, we performed a single-arm, proof-of-concept combination study of these three approaches in ten people with HIV on ART that included (1) therapeutic vaccination with an HIV/Gag conserved element (CE)-targeted DNA+IL-12 prime/MVA boost regimen followed by (2) administration of two bNAbs (10-1074 and VRC07-523LS) and a toll-like receptor 9 (TLR9) agonist (lefitolimod) during ART suppression, followed by (3) repeat bNAb administration at the time of ART interruption. Seven of the ten participants exhibited partial (low viral load set point) or complete (aviremic) post-intervention control after stopping ART, independent of residual bNAb plasma levels. Robust expansion of activated CD8+ T cells early in response to rebounding virus correlated with lower viral load set points. These data suggest that combination immunotherapy approaches might prove effective to induce sustained control of HIV by slowing rebound and improving CD8+ T cell responses, and that these approaches should continue to be optimized.
Collapse
Affiliation(s)
- M.J Peluso
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - D.A Sandel
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - A.N Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, USA
| | - S.J Kim
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - T Dalhuisen
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - H.P Tummala
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, USA
| | - R Tibúrcio
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - L Zemelko
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - G.M Borgo
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - S.S Singh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - K Schwartz
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - M Deswal
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - M.C Williams
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - R Hoh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - M Shimoda
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - S Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - L Serebryannyy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M Khalili
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - E Vendrame
- Gilead Sciences, Inc., Foster City, CA, USA
| | - D SenGupta
- Gilead Sciences, Inc., Foster City, CA, USA
| | - L. S Whitmore
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, USA
| | - J Tisoncik-Go
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, USA
| | - M Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA, USA
- Current affiliation: Department of Microbiology and Immunology, and the Institute on Infectious Diseases, University of Minnesota, Minneapolis, MN, USA
| | - R.A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J.I Mullins
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - B.K Felber
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - G.N Pavlakis
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - J.D Reeves
- Labcorp-Monogram Biosciences, South San Francisco, CA, USA
| | | | - D.V Glidden
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - M.H Spitzer
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
| | - L Gama
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Current affiliation: Instituto Butantan, São Paulo, Brazil
| | - M Caskey
- Department of Clinical Investigation, The Rockefeller University, New York, NY, USA
| | - M.C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - K.W Chew
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - T.J Henrich
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - S.A Yukl
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - L.B Cohn
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - S.G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - R.L Rutishauser
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Garcia-Knight MA, Kelly JD, Lu S, Tassetto M, Goldberg SA, Zhang A, Pineda-Ramirez J, Anglin K, Davidson MC, Chen JY, Fortes-Cobby M, Park S, Martinez A, So M, Donovan A, Viswanathan B, Richardson ET, McIlwain DR, Gaudilliere B, Rutishauser RL, Chenna A, Petropoulos C, Wrin T, Deeks SG, Abedi GR, Saydah S, Martin JN, Briggs Hagen M, Midgley CM, Peluso MJ, Andino R. Circulating neutralizing antibodies and SARS-CoV-2 variant replication following postvaccination infections. JCI Insight 2025; 10:e185953. [PMID: 40059831 PMCID: PMC11949002 DOI: 10.1172/jci.insight.185953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/13/2025] [Indexed: 03/21/2025] Open
Abstract
The effect of preexisting neutralizing antibodies (NAb) on SARS-CoV-2 shedding in postvaccination infection (PVI) is not well understood. We characterized viral shedding longitudinally in nasal specimens in relation to baseline (pre/periinfection) serum NAb titers in 125 participants infected with SARS-CoV-2 variants. Among 68 vaccinated participants, we quantified the effect of baseline NAb titers on maximum viral RNA titers and infectivity duration. Baseline NAbs were higher and targeted a broader range of variants in participants with monovalent ancestral booster vaccinations compared with those with a primary vaccine series. In Delta infections, baseline NAb titers targeting Delta or Wuhan-Hu-1 correlated negatively with maximum viral RNA. Per log10 increase in Delta-targeting baseline NAb IC50, maximum viral load was reduced -2.43 (95% CI: -3.76, -1.11) log10 nucleocapsid copies, and infectious viral shedding was reduced -2.79 (95% CI: -4.99, -0.60) days. Conversely, in Omicron infections (BA.1, BA.2, BA.4, or BA.5), baseline NAb titers against Omicron lineages or Wuhan-Hu-1 did not predict viral outcomes. Our results provide robust estimates of the effect of baseline NAbs on the magnitude and duration of nasal viral replication after PVI (albeit with an unclear effect on transmission) and show how immune escape variants efficiently evade these modulating effects.
Collapse
Affiliation(s)
- Miguel A. Garcia-Knight
- Department of Immunology and Microbiology, UCSF, San Francisco, California, USA
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - J. Daniel Kelly
- Department of Medicine
- Department of Epidemiology and Biostatistics
- Institute for Global Health Sciences, and
- F.I. Proctor Foundation, UCSF, San Francisco, California, USA
- San Francisco VA Medical Center, San Francisco, California, USA
| | - Scott Lu
- Department of Epidemiology and Biostatistics
- Institute for Global Health Sciences, and
| | - Michel Tassetto
- Department of Immunology and Microbiology, UCSF, San Francisco, California, USA
| | - Sarah A. Goldberg
- Department of Epidemiology and Biostatistics
- Institute for Global Health Sciences, and
| | - Amethyst Zhang
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | | | | | | | - Sara Park
- Institute for Global Health Sciences, and
| | | | - Matthew So
- Institute for Global Health Sciences, and
| | - Aidan Donovan
- F.I. Proctor Foundation, UCSF, San Francisco, California, USA
| | | | - Eugene T. Richardson
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, California, USA
| | - Rachel L. Rutishauser
- Division of Experimental Medicine, Department of Medicine, UCSF, San Francisco, California, USA
| | - Ahmed Chenna
- Labcorp - Monogram Biosciences, South San Francisco, California, USA
| | | | - Terri Wrin
- Labcorp - Monogram Biosciences, South San Francisco, California, USA
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases and Global Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Glen R. Abedi
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sharon Saydah
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Melissa Briggs Hagen
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Claire M. Midgley
- Coronavirus and Other Respiratory Viruses Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael J. Peluso
- Division of HIV, Infectious Diseases and Global Medicine, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Raul Andino
- Department of Immunology and Microbiology, UCSF, San Francisco, California, USA
| |
Collapse
|
9
|
Zhu A, Chen Z, Yan Q, Jiang M, Liu X, Li Z, Li N, Tang C, Jian W, He J, Chen L, Cheng J, Chen C, Tang T, Xu Z, Hu Q, Li F, Wang Y, Sun J, Zhuang Z, Wen L, Zhuo J, Liu D, Zhang Y, Huang X, Li S, Zeng Q, Chen F, Zhou L, Liu D, Zhong C, Chen Y, Li S, Liang K, Zhong N, Zhang X, Chen J, Chen X, Xu Y, Zhong N, Zhao J, Zhao J. Robust mucosal SARS-CoV-2-specific T cells effectively combat COVID-19 and establish polyfunctional resident memory in patient lungs. Nat Immunol 2025; 26:459-472. [PMID: 39875584 PMCID: PMC11876067 DOI: 10.1038/s41590-024-02072-9] [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: 06/04/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025]
Abstract
Mucosal antigen-specific T cells are pivotal for pathogen clearance and immune modulation in respiratory infections. Dysregulated T cell responses exacerbate coronavirus disease 2019 severity, marked by cytokine storms and respiratory failure. Despite extensive description in peripheral blood, the characteristics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells in the lungs remain elusive. Here we conducted integrated single-cell profiling of SARS-CoV-2-specific T cells in 122 bronchoalveolar lavage fluid (BALF) and 280 blood samples from 159 patients, including 27 paired BALF and blood samples from 24 patients. SARS-CoV-2-specific T cells were robustly elicited in BALF irrespective of prior vaccination, correlating with diminished viral loads, lessened systemic inflammation and improved respiratory function. SARS-CoV-2-specific T cells in BALF exhibited profound activation, along with proliferative and multi-cytokine-producing capabilities and a glycolysis-driven metabolic signature, which were distinct from those observed in peripheral blood mononuclear cells. After viral clearance, these specific T cells maintained a polyfunctional tissue-resident memory phenotype, highlighting their critical roles in infection control and long-term protection.
Collapse
Grants
- the National Key R&D Program of China (2023YFC2306400 to JC.Z., 2023YFC3041700 to JC.Z.), R&D Program of Guangzhou Laboratory (SRPG22-006 to JC.Z.), National Natural Science Foundation of China (82495200 to JC.Z., 82495203 to JC.Z., 82025001 to JC.Z.)
- R&D Program of Guangzhou Laboratory (EKPG21-29 to A.Z.),National Natural Science Foundation of China (82201933 to A.Z.), China Postdoctoral Science Foundation (2022M710892 to A.Z.), State Key Laboratory of Respiratory Diseases Funded Program (SKLRD-Z-202330 to A.Z.).
- National Natural Science Foundation of China (82201932 to Q.Y.),China Postdoctoral Science Foundation (2022M710891 to Q.Y.), State Key Laboratory of Respiratory Diseases Funded Program (SKLRD-Z-202324 to Q.Y).
- State Key Laboratory of Respiratory Diseases Funded Program (SKLRD-Z-202304 to Q.H.).
- National Key R&D Program of China (2021YFC0864500 to Y.X.), R&D Program of Guangzhou Laboratory (SRPG23-001 to Y.X.)
- Guangdong Basic and Applied Basic Research Foundation (2021B1515130005 to JX.Z.)
Collapse
Affiliation(s)
- Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qihong Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mei Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuesong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhengtu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Na Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Chunli Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhua Jian
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Lan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Jinling Cheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Canjie Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tian Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiwei Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liyan Wen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianfen Zhuo
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Donglan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanjun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaofang Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suxiang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiuhui Zeng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangli Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liang Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongdong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changhao Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kangli Liang
- Guangdong Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, China
| | - Na Zhong
- Shenzhen Peacock Biotechnology Co. Ltd, Shenzhen, China
| | - Xinmei Zhang
- Shenzhen Peacock Biotechnology Co. Ltd, Shenzhen, China
| | - Jiekai Chen
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaobo Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yonghao Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
10
|
Kalimuddin S, Tham CYL, Chan YFZ, Hang SK, Kunasegaran K, Chia A, Chan CYY, Ng DHL, Sim JXY, Tan HC, Syenina A, Ngoh AQ, Hamis NZ, Chew V, Leong YS, Yee JX, Low JG, Chan KR, Ong EZ, Bertoletti A, Ooi EE. Vaccine-induced T cell responses control Orthoflavivirus challenge infection without neutralizing antibodies in humans. Nat Microbiol 2025; 10:374-387. [PMID: 39794472 PMCID: PMC11790491 DOI: 10.1038/s41564-024-01903-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 12/04/2024] [Indexed: 01/13/2025]
Abstract
T cells have been identified as correlates of protection in viral infections. However, the level of vaccine-induced T cells needed and the extent to which they alone can control acute viral infection in humans remain uncertain. Here we conducted a double-blind, randomized controlled trial involving vaccination and challenge in 33 adult human volunteers, using the live-attenuated yellow fever (YF17D) and chimeric Japanese encephalitis-YF17D (JE/YF17D) vaccines. Both Orthoflavivirus vaccines share T cell epitopes but have different neutralizing antibody epitopes. The primary objective was to assess the extent to which vaccine-induced T cell responses, independent of neutralizing antibodies, were able to reduce post-challenge viral RNAaemia levels. Secondary objectives included an assessment of surrogate measures of viral control, including post-challenge antibody titres and symptomatic outcomes. YF17D vaccinees had reduced levels of JE/YF17D challenge viraemia, compared with those without previous YF17D vaccination (mean log10(area under the curve genome copies per ml): 2.23 versus 3.22; P = 0.039). Concomitantly, YF17D vaccinees had lower post-JE/YF17D challenge antibody titres that reduced JE virus plaque number by 50%, or PRNT50 (mean log10(PRNT50 titre): 1.87 versus 2.5; P < 0.0001) and symptomatic rates (6% (n = 1/16) versus 53% (n = 9/17), P = 0.007). There were no unexpected safety events. Importantly, after challenge infection, several vaccinees had undetectable viraemia and no seroconversion, even in the absence of neutralizing antibodies. Indeed, high vaccine-induced T cell responses, specifically against the capsid protein, were associated with a level of viral control conventionally interpreted as sterilizing immunity. Our findings reveal the importance of T cells in controlling acute viral infection and suggests a potential correlate of protection against orthoflaviviral infections. ClinicalTrials.gov registration: NCT05568953 .
Collapse
Affiliation(s)
- Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore.
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
| | - Christine Y L Tham
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Yvonne F Z Chan
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Shou Kit Hang
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Kamini Kunasegaran
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Adeline Chia
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Candice Y Y Chan
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Dorothy H L Ng
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Jean X Y Sim
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
- Department of Infection Prevention and Epidemiology, Singapore General Hospital, Singapore, Singapore
| | - Hwee-Cheng Tan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Ayesa Syenina
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - An Qi Ngoh
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Noor Zayanah Hamis
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Valerie Chew
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Yan Shan Leong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Jia Xin Yee
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Jenny G Low
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Kuan Rong Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Eugenia Z Ong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Antonio Bertoletti
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Singapore Immunology Network, A*STAR Singapore, Singapore, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
- Department of Translational Clinical Research, Singapore General Hospital, Singapore, Singapore.
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
11
|
Chan L, Pinedo K, Stabile MA, Hamlin RE, Pienkos SM, Ratnasiri K, Yang S, Blomkalns AL, Nadeau KC, Pulendran B, O'Hara R, Rogers AJ, Holmes SP, Blish CA. Prior vaccination prevents overactivation of innate immune responses during COVID-19 breakthrough infection. Sci Transl Med 2025; 17:eadq1086. [PMID: 39879318 DOI: 10.1126/scitranslmed.adq1086] [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: 05/03/2024] [Revised: 09/10/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025]
Abstract
At this stage in the COVID-19 pandemic, most infections are "breakthrough" infections that occur in individuals with prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure. To refine long-term vaccine strategies against emerging variants, we examined both innate and adaptive immunity in breakthrough infections. We performed single-cell transcriptomic, proteomic, and functional profiling of primary and breakthrough infections to compare immune responses from unvaccinated and vaccinated individuals during the SARS-CoV-2 Delta wave. Breakthrough infections were characterized by a less activated transcriptomic profile in monocytes and natural killer cells, with induction of pathways limiting monocyte migratory potential and natural killer cell proliferation. Furthermore, we observed a female-specific increase in transcriptomic and proteomic activation of multiple innate immune cell subsets during breakthrough infections. These insights suggest that prior SARS-CoV-2 vaccination prevents overactivation of innate immune responses during breakthrough infections with discernible sex-specific patterns and underscore the potential of harnessing vaccines in mitigating pathologic immune responses resulting from overactivation.
Collapse
Affiliation(s)
- Leslie Chan
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kassandra Pinedo
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mikayla A Stabile
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca E Hamlin
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shaun M Pienkos
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kalani Ratnasiri
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Samuel Yang
- Department of Emergency Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andra L Blomkalns
- Department of Emergency Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kari C Nadeau
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ruth O'Hara
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Angela J Rogers
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
12
|
Noureddine M, Chang LA, El Ayache F, Laghlali G, Burgess E, Gruneberg L, Warang P, Jiang K, Nijhuis H, Coughlan L, Diego JGB, Park S, Levican J, Schotsaert M. Muscle macrophage regenerative response after squalene-adjuvanted influenza vaccination drives Th2-skewed response and is reduced with age. RESEARCH SQUARE 2025:rs.3.rs-5760877. [PMID: 39975920 PMCID: PMC11838721 DOI: 10.21203/rs.3.rs-5760877/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Squalene-based adjuvants like MF59, and its research alternative AddaVax, induce transient muscle injury, but their working mechanisms downstream of muscle injury remain unclear. We show that an AddaVax-adjuvanted quadrivalent inactivated influenza virus vaccine (QIV) intramuscular injection triggers muscle regeneration-like immune processes and increases CX3CR1+Ly6C+ macrophages in the muscle and inguinal lymph nodes by day 4 post-injection. This leads to a Th2 skewed vaccine response with higher levels of vaccine specific IgG1 titers, and Th2-associated cytokines in the lungs 5 days after subsequent influenza viral challenge. In aged mice, the macrophage recruitment and polarization is diminished, which is consistent with age-associated muscle mass loss, reflecting the age-related decline in muscle regeneration. Unlike young mice, aged mice exhibit a reduction in magnitude and skewing of AddaVax-mediated Th2 responses to QIV. We found that adoptive transfer of bone marrow-derived macrophages derived from young mice into aged mice at the moment of vaccination leads to their infiltration into the injected muscle, where they collect vaccine antigens, drain to the lymph node, and enhance the Th2 response, recapitulating the young host response but in an older host. However, rescuing the Th2-skewing effects of AddaVax alone was not sufficient to enhance protection against mismatched subsequent influenza viral infection in aged mice, suggesting additional factors at play in the diminished vaccine response in aged hosts. This underscores the importance of the macrophage-driven muscle regenerative response in the mechanism of action for squalene-based adjuvants like AddaVax and emphasizes the need to study how muscle damage and regenerative pathways in intramuscular vaccine responses contribute to vaccine effectiveness.
Collapse
Affiliation(s)
- Moataz Noureddine
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Farah El Ayache
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gabriel Laghlali
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Eleanor Burgess
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Leonie Gruneberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kaijun Jiang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Haye Nijhuis
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Juan Garcia-Bernalt Diego
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Seokchan Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jorge Levican
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
13
|
Liu J, Wang L, Kurtesi A, Budylowski P, Potts KG, Menon H, Tan Y, Samaan P, Liu X, Wang Y, Hu Q, Samson R, Qi F, Evseev D, John C, Ellestad KK, Fan Y, Budiman F, Tohan ER, Udayakumar S, Yang J, Marcusson EG, Gingras AC, Mahoney DJ, Ostrowski MA, Martin-Orozco N. A bivalent COVID-19 mRNA vaccine elicited broad immune responses and protection against Omicron subvariants infection. NPJ Vaccines 2025; 10:4. [PMID: 39788981 PMCID: PMC11718203 DOI: 10.1038/s41541-025-01062-8] [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: 04/04/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
Continuously emerging SARS-CoV-2 Omicron subvariants pose a threat thwarting the effectiveness of approved COVID-19 vaccines. Especially, the protection breadth and degree of these vaccines against antigenically distant Omicron subvariants is unclear. Here, we report the immunogenicity and efficacy of a bivalent mRNA vaccine, PTX-COVID19-M1.2 (M1.2), which encodes native spike proteins from Wuhan-Hu-1 (D614G) and Omicron BA.2.12.1, in mouse and hamster models. Both primary series and booster vaccination using M1.2 elicited potent and broad nAbs against Wuhan-Hu-1 (D614G) and some Omicron subvariants. Strong spike-specific T cell responses against Wuhan-Hu-1 and Omicron subvariants, including JN.1, were also induced. Vaccination with M1.2 protected animals from Wuhan-Hu-1 and multiple Omicron subvariants challenges. Interestingly, protection against XBB.1.5 lung infection did not correlate with nAb levels. These results indicate that M1.2 generated a broadly protective immune response against antigenically distant Omicron subvariants, and spike-specific T cells probably contributed to the breadth of the protection.
Collapse
Affiliation(s)
- Jun Liu
- Providence Therapeutics Holdings, Inc., Calgary, Canada.
| | - Li Wang
- Everest Medicines, Shanghai, China
| | - Alexandra Kurtesi
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| | - Patrick Budylowski
- Department of Medicine, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Kyle G Potts
- Riddell Center for Cancer Immunotherapy, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Molecular Biology and Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Haritha Menon
- Providence Therapeutics Holdings, Inc., Calgary, Canada
| | - Yilin Tan
- Providence Therapeutics Holdings, Inc., Calgary, Canada
| | - Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | | | | - Queenie Hu
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| | - Reuben Samson
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| | - Freda Qi
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| | - Danyel Evseev
- Riddell Center for Cancer Immunotherapy, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Molecular Biology and Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Cini John
- Riddell Center for Cancer Immunotherapy, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Molecular Biology and Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Kristofor K Ellestad
- Riddell Center for Cancer Immunotherapy, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Molecular Biology and Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Yue Fan
- Everest Medicines, Shanghai, China
| | - Frans Budiman
- Department of Medicine, University of Toronto, Toronto, Canada
| | | | - Suji Udayakumar
- Department of Medicine, University of Toronto, Toronto, Canada
| | | | | | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| | - Douglas J Mahoney
- Riddell Center for Cancer Immunotherapy, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Molecular Biology and Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Mario A Ostrowski
- Department of Medicine, University of Toronto, Toronto, Canada.
- Institute of Medical Science, University of Toronto, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Department of Immunology, University of Toronto, Toronto, Canada.
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.
| | | |
Collapse
|
14
|
da Silva Antunes R, Fajardo-Rosas V, Yu ED, Gálvez RI, Abawi A, Alexandar Escarrega E, Martínez-Pérez A, Johansson E, Goodwin B, Frazier A, Dan JM, Crotty S, Seumois G, Weiskopf D, Vijayanand P, Sette A. Evolution of SARS-CoV-2 T cell responses as a function of multiple COVID-19 boosters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631842. [PMID: 39829792 PMCID: PMC11741356 DOI: 10.1101/2025.01.08.631842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The long-term effects of repeated COVID-19 vaccinations on adaptive immunity remain incompletely understood. Here, we conducted a comprehensive three-year longitudinal study examining T cell and antibody responses in 78 vaccinated individuals without reported symptomatic infections. We observed distinct dynamics in Spike-specific humoral and cellular immune responses across multiple vaccine doses. While antibody titers incrementally increased and stabilized with each booster, T cell responses rapidly plateaued, maintaining remarkable stability across CD4+ and CD8+ subsets. Notably, approximately 30% of participants showed CD4+ T cell reactivity to non-Spike antigens, consistent with asymptomatic infections. Single-cell RNA sequencing revealed a diverse landscape of Spike-specific T cell phenotypes, with no evidence of increased exhaustion or significant functional impairment. However, qualitative changes were observed in individuals with evidence of asymptomatic infection, exhibiting unique immunological characteristics, including increased frequencies of Th17-like CD4+ T cells and GZMKhi/IFNR CD8+ T cell subsets. Remarkably, repeated vaccinations in this group were associated with a progressive increase in regulatory T cells, potentially indicating a balanced immune response that may mitigate immunopathology. By regularly stimulating T cell memory, boosters contribute to a stable and enhanced immune response, which may provide better protection against symptomatic infections.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- These authors contributed equally
| | - Vicente Fajardo-Rosas
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Bioinformatics and Systems Biology Graduate Program; University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Rosa Isela Gálvez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Adam Abawi
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - E. Alexandar Escarrega
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Amparo Martínez-Pérez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Emil Johansson
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Benjamin Goodwin
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jennifer M. Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Grégory Seumois
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Pandurangan Vijayanand
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Senior authorship
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Senior authorship
- Lead contact
| |
Collapse
|
15
|
Packer CH, Jasset O, Hanniford N, Brigida S, Demidkin S, Perlis RH, Edlow AG, Shook LL. Maternal-fetal cytokine profiles in acute SARS-CoV-2 "breakthrough" infection after COVID-19 vaccination. Front Immunol 2025; 15:1506203. [PMID: 39845965 PMCID: PMC11750656 DOI: 10.3389/fimmu.2024.1506203] [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: 10/07/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Objective Vaccination is protective against severe COVID-19 disease, yet whether vaccination reduces COVID-19-associated inflammation in pregnancy has not been established. The objective of this study is to characterize maternal and cord cytokine profiles of acute SARS-CoV-2 "breakthrough" infection (BTI) after vaccination, compared with unvaccinated infection and uninfected controls. Study design 66 pregnant individuals enrolled in the MGH COVID-19 biorepository (March 2020-April 2022) were included. Maternal sera were collected from 26 unvaccinated and 21 vaccinated individuals with acute SARS-CoV-2 infection. Cord sera were collected at delivery. Maternal and cord sera from 19 term dyads without current or prior SARS-CoV-2 infection were analyzed as controls. Cytokines were quantified using the Human Inflammation 20-Plex ProcartaPlex assay. Results There was a significantly higher incidence of severe/critical maternal illness in unvaccinated pregnant individuals with SARS-CoV-2 compared to vaccinated (10/26 (38%) vs. 0/21 (0%), p<0.01). Significantly higher maternal levels of TNFα and CD62P were observed in vaccinated individuals with SARS-CoV-2 BTI compared with unvaccinated individuals with infection (p<0.05). Network correlation analyses revealed a distinct maternal cytokine response to SARS-CoV-2 in vaccinated vs unvaccinated individuals. Neither unvaccinated nor vaccinated SARS-CoV-2 infection resulted in elevated cord cytokines compared to controls. Multivariate analyses demonstrate distinct maternal and cord cytokine profiles in the setting of maternal SARS-CoV-2 at delivery. Conclusion Vaccination was associated with higher maternal cytokine levels during acute SARS-CoV-2 infection compared to unvaccinated infection, which may reflect vaccine-mediated priming of the immune system. A fetal inflammatory response specific to maternal SARS-CoV-2 infection was not observed.
Collapse
Affiliation(s)
- Claire H. Packer
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Olyvia Jasset
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Nikolina Hanniford
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sara Brigida
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Stepan Demidkin
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Roy H. Perlis
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Andrea G. Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| | - Lydia L. Shook
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
16
|
Mahrokhian SH, Tostanoski LH, Vidal SJ, Barouch DH. COVID-19 vaccines: Immune correlates and clinical outcomes. Hum Vaccin Immunother 2024; 20:2324549. [PMID: 38517241 PMCID: PMC10962618 DOI: 10.1080/21645515.2024.2324549] [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: 01/24/2024] [Accepted: 02/24/2024] [Indexed: 03/23/2024] Open
Abstract
Severe disease due to COVID-19 has declined dramatically as a result of widespread vaccination and natural immunity in the population. With the emergence of SARS-CoV-2 variants that largely escape vaccine-elicited neutralizing antibody responses, the efficacy of the original vaccines has waned and has required vaccine updating and boosting. Nevertheless, hospitalizations and deaths due to COVID-19 have remained low. In this review, we summarize current knowledge of immune responses that contribute to population immunity and the mechanisms how vaccines attenuate COVID-19 disease severity.
Collapse
Affiliation(s)
- Shant H. Mahrokhian
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Samuel J. Vidal
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
17
|
Ramirez SI, Lopez PG, Faraji F, Parikh UM, Heaps A, Ritz J, Moser C, Eron JJ, Wohl D, Currier J, Daar ES, Greninger A, Klekotka P, Grifoni A, Weiskopf D, Sette A, Peters B, Hughes MD, Chew KW, Smith DM, Crotty S. Early antiviral CD4+ and CD8+ T cells are associated with upper airway clearance of SARS-CoV-2. JCI Insight 2024; 9:e186078. [PMID: 39704169 DOI: 10.1172/jci.insight.186078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/25/2024] [Indexed: 12/21/2024] Open
Abstract
T cells are involved in protective immunity against numerous viral infections. Data regarding functional roles of human T cells in SARS-CoV-2 (SARS2) viral clearance in primary COVID-19 are limited. To address this knowledge gap, we assessed samples for associations between SARS2 upper respiratory tract viral RNA levels and early virus-specific adaptive immune responses for 95 unvaccinated clinical trial participants with acute primary COVID-19 aged 18-86 years old, approximately half of whom were considered at high risk for progression to severe COVID-19. Functionality and magnitude of acute SARS2-specific CD4+ and CD8+ T cell responses were evaluated, in addition to antibody responses. Most individuals with acute COVID-19 developed SARS2-specific T cell responses within 6 days of COVID-19 symptom onset. Early CD4+ T cell and CD8+ T cell responses were polyfunctional, and both strongly associated with reduced upper respiratory tract SARS2 viral RNA, independent of neutralizing antibody titers. Overall, these findings provide evidence for protective roles for circulating SARS2-specific CD4+ and CD8+ T cells during acute COVID-19.
Collapse
Affiliation(s)
- Sydney I Ramirez
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Paul G Lopez
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Farhoud Faraji
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Otolaryngology-Head and Neck Surgery, UCSD, La Jolla, California, USA
| | - Urvi M Parikh
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy Heaps
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Justin Ritz
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Carlee Moser
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Joseph J Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - David Wohl
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Judith Currier
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Eric S Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Alex Greninger
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA
| | | | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Daniela Weiskopf
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Michael D Hughes
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kara W Chew
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Davey M Smith
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Shane Crotty
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| |
Collapse
|
18
|
Simayi A, Chen Y, Chu J, Yu H, Zhang S, Bao C, Zhu F, Jin H, Qin Y, Zhen Q, Liu Y, Zhu L. Ad5-nCoV boosted vaccine and reinfection-induced memory T/B cell responses and humoral immunity to SARS-CoV-2: based on two prospective cohorts. Emerg Microbes Infect 2024; 13:2412619. [PMID: 39360715 PMCID: PMC11529888 DOI: 10.1080/22221751.2024.2412619] [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: 06/18/2024] [Revised: 09/04/2024] [Accepted: 09/30/2024] [Indexed: 10/04/2024]
Abstract
Here, we regularly followed two SARS-CoV-2 infected cohorts to investigate the combined effects of neutralizing antibodies (NAbs) and B and T cell profiles during the convalescent period. Ten infected participants in December 2022 were selected to assess the effects of an inhaled adenovirus type 5 vectored COVID-19 vaccine (Ad5-nCoV) booster on B cells and humoral immunity in the first cohort. To evaluate T cell responses, eight primary and 20 reinfection participants were included in the second cohort. Blood samples from all 38 participants were collected at 1-, 2-, and 6-months post-infection. In the first cohort, eighteen monoclonal antibodies (mAbs) with neutralizing activity from memory B cells (MBC) against SARS-CoV-2 mutants were obtained by high throughput single-B-cell cloning method, which lasted from 1- month to 6- month post infection. The overall number of mAbs from MBC in the boosted immunization group was higher than that in the nonboosted immunization group at 2-, and 6-months post-infection. In the second cohort, circulating T follicular helper cells (cTfh) and AIM + CD4 + T cells increased over time in the reinfection group (P < 0.05). In both cohorts, serum NAb titers showed significant immune escape, while cTfh and AIM + CD4 + T cells in the second cohort essentially showed no immune escape to new strains (including XBB, EG.5). AIM + CD4 + T cells against BA.5 and EG.5 were strongly negatively correlated with the time to viral clearance in the reinfected group at 6-months post-infection. We comprehensively assessed the ability of the SARS-CoV-2 boosted immunization and reinfection-induced generation of T/B cell immune memories in preventing reinfection.
Collapse
Affiliation(s)
- Aidibai Simayi
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, People’s Republic of China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People’s Republic of China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing
| | - Jinjin Chu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
| | - Huiyan Yu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
| | - Shihan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, People’s Republic of China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People’s Republic of China
| | - Changjun Bao
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
- Jiangsu Province Engineering Research Center of Health Emergency, Nanjing, People’s Republic of China
| | - Fengcai Zhu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
- Key Laboratory of Infectious Diseases, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Hui Jin
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, People’s Republic of China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People’s Republic of China
| | - Yuanfang Qin
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
| | - Qian Zhen
- Department of Acute Infectious Disease Control and Prevention, Changzhou Center for Disease Control and Prevention, Changzhou, People’s Republic of China
| | - Yong Liu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing
| | - Liguo Zhu
- Department of Acute Infectious Disease Control and Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
- Key Laboratory of Infectious Diseases, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People’s Republic of China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
19
|
Torán-Monserrat P, Lamonja-Vicente N, Costa-Garrido A, Carrasco-Ribelles LA, Quirant B, Boigues M, Molina X, Chacón C, Dacosta-Aguayo R, Arméstar F, Martínez Cáceres EM, Prado JG, Violán C. SARS-CoV-2 Infection Risk by Vaccine Doses and Prior Infections Over 24 Months: ProHEpiC-19 Longitudinal Study. JMIR Public Health Surveill 2024; 10:e56926. [PMID: 39648969 PMCID: PMC11606241 DOI: 10.2196/56926] [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: 01/31/2024] [Revised: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 12/10/2024] Open
Abstract
Background As the vaccination campaign against COVID-19 progresses, it becomes crucial to comprehend the lasting effects of vaccination on safeguarding against new infections or reinfections. Objective This study aimed to assess the risk of new SARS-CoV-2 infections based on the number of vaccine doses, prior infections, and other clinical characteristics. Methods We defined a cohort of 800 health care workers in a 24-month study (March 2020 to December 2022) in northern Barcelona to determine new infections by SARS-CoV-2. We used extended Cox models, specifically Andersen-Gill (AG) and Prentice-Williams-Peterson, and we examined the risk of new infections. The AG model incorporated variables such as sex, age, job title, number of chronic conditions, vaccine doses, and prior infections. Additionally, 2 Prentice-Williams-Peterson models were adjusted, one for those individuals with no or 1 infection and another for those with 2 or 3 infections, both with the same covariates as the AG model. Results The 800 participants (n=605, 75.6% women) received 1, 2, 3, and 4 doses of the vaccine. Compared to those who were unvaccinated, the number of vaccine doses significantly reduced (P<.001) the risk of infection by 66%, 81%, 89%, and 99%, respectively. Unit increase in the number of prior infections reduced the risk of infection by 75% (P<.001). When separating individuals by number of previous infections, risk was significantly reduced for those with no or 1 infection by 61% (P=.02), and by 88%, 93%, and 99% (P<.001) with 1, 2, 3, or 4 doses, respectively. In contrast, for those with 2 or 3 previous infections, the reduction was only significant with the fourth dose, at 98% (P<.001). The number of chronic diseases only increased the risk by 28%-31% (P<.001) for individuals with 0-1 previous infections. Conclusions The study suggests that both prior infections and vaccination status significantly contribute to SARS-CoV-2 immunity, supporting vaccine effectiveness in reducing risk of reinfection for up to 24 months after follow-up from the onset of the pandemic. These insights contribute to our understanding of long-term immunity dynamics and inform strategies for mitigating the impact of COVID-19.
Collapse
Affiliation(s)
- Pere Torán-Monserrat
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mare de Déu de Guadalupe, 2, Mataró, 08303, Spain, 34 7415338
- Germans Trias i Pujol Research Institute, Badalona, Spain
- Department of Medicine, Faculty of Medicine, Universitat de Girona, Girona, Spain
- Multidisciplinary Research Group in Health and Society (2021-SGR-0148), Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mataró, Spain
| | - Noemí Lamonja-Vicente
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mare de Déu de Guadalupe, 2, Mataró, 08303, Spain, 34 7415338
| | - Anna Costa-Garrido
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mare de Déu de Guadalupe, 2, Mataró, 08303, Spain, 34 7415338
| | - Lucía A Carrasco-Ribelles
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mare de Déu de Guadalupe, 2, Mataró, 08303, Spain, 34 7415338
- Grup de REcerca en Impacte de les Malalties Cròniques i les seves Trajectòries (2021 SGR 01537), Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
| | - Bibiana Quirant
- Immunology Department, Federation of Clinical Immunology Societies Center of Excellence, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Immunology Division, Laboratori Clinic Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Marc Boigues
- Immunology Department, Federation of Clinical Immunology Societies Center of Excellence, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Immunology Division, Laboratori Clinic Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Xaviera Molina
- Programa de Màster en Salud Pública, Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Epidemiología y Políticas de Salud, Universidad del Desarrollo, Santiago de Chile, Chile
| | - Carla Chacón
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mare de Déu de Guadalupe, 2, Mataró, 08303, Spain, 34 7415338
| | - Rosalia Dacosta-Aguayo
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mare de Déu de Guadalupe, 2, Mataró, 08303, Spain, 34 7415338
| | - Fernando Arméstar
- Intensive Care Unit, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Eva María Martínez Cáceres
- Immunology Department, Federation of Clinical Immunology Societies Center of Excellence, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Immunology Division, Laboratori Clinic Metropolitana Nord, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Julia G Prado
- Germans Trias i Pujol Research Institute, Badalona, Spain
- IrsiCaixa-AIDS Research Institute, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Concepción Violán
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Mare de Déu de Guadalupe, 2, Mataró, 08303, Spain, 34 7415338
- Grup de REcerca en Impacte de les Malalties Cròniques i les seves Trajectòries (2021 SGR 01537), Institut Universitari d’Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Grup de Recerca en Impacte de les Malalties Cròniques i les seves Trajectòries (2021-SGR-01537), Germans Trias i Pujol Research Institute, Badalona, Spain
- Red de Investigación en Cronicidad, Atención Primaria y Prevención y Promoción de la Salut, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
20
|
Byrne C, Schiffer JT. Ensemble modeling of SARS-CoV-2 immune dynamics in immunologically naïve rhesus macaques predicts that potent, early innate immune responses drive viral elimination. Front Immunol 2024; 15:1426016. [PMID: 39575237 PMCID: PMC11578959 DOI: 10.3389/fimmu.2024.1426016] [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: 04/30/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction An unprecedented breadth of longitudinal viral and multi-scale immunological data has been gathered during SARS-CoV-2 infection. However, due to the high complexity, non-linearity, multi-dimensionality, mixed anatomic sampling, and possible autocorrelation of available immune data, it is challenging to identify the components of the innate and adaptive immune response that drive viral elimination. Novel mathematical models and analytical approaches are required to synthesize contemporaneously gathered cytokine, transcriptomic, flow cytometry, antibody response, and viral load data into a coherent story of viral control, and ultimately to discriminate drivers of mild versus severe infection. Methods We investigated a dataset describing innate, SARS-CoV-2 specific T cell, and antibody responses in the lung during early and late stages of infection in immunologically naïve rhesus macaques. We used multi-model inference and ensemble modeling approaches from ecology and weather forecasting to compare and combine various competing models. Results and discussion Model outputs suggest that the innate immune response plays a crucial role in controlling early infection, while SARS-CoV-2 specific CD4+ T cells correspond to later viral elimination, and anti-spike IgG antibodies do not impact viral dynamics. Among the numerous genes potentially contributing to the innate response, we identified IFI27 as most closely linked to viral load decline. A 90% knockdown of the innate response from our validated model resulted in a ~10-fold increase in peak viral load during infection. Our approach provides a novel methodological framework for future analyses of similar complex, non-linear multi-component immunologic data sets.
Collapse
Affiliation(s)
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center,
Seattle, WA, United States
| |
Collapse
|
21
|
Altmann DM, Boyton RJ. Protective immunity to repeated COVID-19 breakthrough infections. Clin Immunol 2024; 268:110374. [PMID: 39357633 DOI: 10.1016/j.clim.2024.110374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Daniel M Altmann
- Departments of Immunology and Inflammation, Faculty of Medicine, Imperial College London, UK.
| | - Rosemary J Boyton
- Departments of Infectious Disease, Faculty of Medicine, Imperial College London, UK
| |
Collapse
|
22
|
Sanchez S, Dangi T, Awakoaiye B, Lew MH, Irani N, Fourati S, Penaloza-MacMaster P. Delayed reinforcement of costimulation improves the efficacy of mRNA vaccines in mice. J Clin Invest 2024; 134:e183973. [PMID: 39432667 PMCID: PMC11645141 DOI: 10.1172/jci183973] [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: 06/26/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
mRNA vaccines have demonstrated efficacy during the COVID-19 pandemic and are now being investigated for multiple diseases. However, concerns linger about the durability of immune responses, and the high incidence of breakthrough infections among vaccinated individuals highlights the need for improved mRNA vaccines. In this study, we investigated the effects of reinforcing costimulation via 4-1BB, a member of the TNF receptor superfamily, on immune responses elicited by mRNA vaccines. We first immunized mice with mRNA vaccines, followed by treatment with 4-1BB costimulatory antibodies to reinforce the 4-1BB pathway at different time points after vaccination. Consistent with prior studies, reinforcing 4-1BB costimulation on the day of vaccination did not result in a substantial improvement in vaccine responses. However, reinforcing 4-1BB costimulation on day 4 after vaccination, when 4-1BB expression levels were highest, resulted in a profound improvement in CD8+ T cell responses associated with enhanced protection against pathogen challenges. A similar clinical benefit was observed in a therapeutic cancer vaccine model. We also report time-dependent effects with OX40, another costimulatory molecule of the TNF receptor superfamily. These findings demonstrate that delayed reinforcement of costimulation may exert an immunologic benefit, providing insights for the development of more effective mRNA vaccines for infectious diseases and cancer.
Collapse
MESH Headings
- mRNA Vaccines/administration & dosage
- mRNA Vaccines/immunology
- Immunogenicity, Vaccine
- Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
- Vaccination/methods
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- OX40 Ligand/agonists
- OX40 Ligand/immunology
- OX40 Ligand/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Differentiation/immunology
- Mice, Inbred C57BL
- Animals
- Mice
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Immunization, Secondary/methods
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Male
- Female
Collapse
Affiliation(s)
| | | | | | | | | | - Slim Fourati
- Department of Medicine, Division of Allergy and Immunology, Feinberg School of Medicine and Center for Human Immunobiology, Northwestern University, Chicago, Illinois, USA
| | | |
Collapse
|
23
|
Zhao XJ, Li XL, Zhang S, Chen JJ, Zhao WC, Wu NN, Wang RJ, Xu Q, Lv CL, Jiang BG, Wang GL, Fang LQ. Dynamic changes of neutralizing antibody and memory T cell responses six months post Omicron XBB reinfection. Front Immunol 2024; 15:1477721. [PMID: 39434881 PMCID: PMC11491401 DOI: 10.3389/fimmu.2024.1477721] [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: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction With the continued prevalence of COVID-19, repeated infection caused by SARS-CoV-2 has become common. However, studies on immune persistence post Omicron XBB reinfection are limited. Methods We prospectively studied the durability and cross-reactivity of neutralizing antibodies (NAbs) and T cell responses among 20 subjects who suffered Omicron BA.5 infection with or without Omicron XBB reinfection over 6-month through the pseudovirus neutralization test and the fluorospot assay. Results NAbs against EG.5.1, BA.2.86, and JN.1 subvariants were decreased and undetectable at 6-month post Omicron BA.5 infection, while those elicited by Omicron XBB reinfection were significant increased and remained detectable against all detected variants within 6-month. Furthermore, in subjects with Omicron XBB reinfection, memory T cell responses could cross-recognized wild-type and Omicron spike peptides and reached peak at 3-month. Interestingly, comparable robust T cell responses were observed among non-seroconverted subjects post Omicron XBB exposure. Conclusion Though the NAbs against various emerging Omicron subvariants elicited by Omicron XBB reinfection can persist for at least 6-month, the HCWs should strengthen personal protection and timely be immunized with updated vaccines upon current circulating variants or conserved T epitope.
Collapse
Affiliation(s)
- Xin-Jing Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Xin-Lou Li
- Department of Medical Research, Key Laboratory of Environmental Sense Organ Stress and Health of the Ministry of Environmental Protection, the Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Sheng Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Jin-Jin Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Wei-Chao Zhao
- Department of Respiratory Medicine, the Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Na-Na Wu
- Department of Respiratory Medicine, the Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Rui-Juan Wang
- Department of Respiratory Medicine, the Ninth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qiang Xu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Chen-Long Lv
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Bao-Gui Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Guo-Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Li-Qun Fang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| |
Collapse
|
24
|
Gujar S, Pol JG, Kumar V, Lizarralde-Guerrero M, Konda P, Kroemer G, Bell JC. Tutorial: design, production and testing of oncolytic viruses for cancer immunotherapy. Nat Protoc 2024; 19:2540-2570. [PMID: 38769145 DOI: 10.1038/s41596-024-00985-1] [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: 05/10/2023] [Accepted: 02/12/2024] [Indexed: 05/22/2024]
Abstract
Oncolytic viruses (OVs) represent a novel class of cancer immunotherapy agents that preferentially infect and kill cancer cells and promote protective antitumor immunity. Furthermore, OVs can be used in combination with established or upcoming immunotherapeutic agents, especially immune checkpoint inhibitors, to efficiently target a wide range of malignancies. The development of OV-based therapy involves three major steps before clinical evaluation: design, production and preclinical testing. OVs can be designed as natural or engineered strains and subsequently selected for their ability to kill a broad spectrum of cancer cells rather than normal, healthy cells. OV selection is further influenced by multiple factors, such as the availability of a specific viral platform, cancer cell permissivity, the need for genetic engineering to render the virus non-pathogenic and/or more effective and logistical considerations around the use of OVs within the laboratory or clinical setting. Selected OVs are then produced and tested for their anticancer potential by using syngeneic, xenograft or humanized preclinical models wherein immunocompromised and immunocompetent setups are used to elucidate their direct oncolytic ability as well as indirect immunotherapeutic potential in vivo. Finally, OVs demonstrating the desired anticancer potential progress toward translation in patients with cancer. This tutorial provides guidelines for the design, production and preclinical testing of OVs, emphasizing considerations specific to OV technology that determine their clinical utility as cancer immunotherapy agents.
Collapse
Affiliation(s)
- Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Jonathan G Pol
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
| | - Vishnupriyan Kumar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Manuela Lizarralde-Guerrero
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
- Ecole Normale Supérieure de Lyon, Lyon, France
| | - Prathyusha Konda
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Guido Kroemer
- INSERM, U1138, Paris, France.
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
- Université Paris Cité, Paris, France.
- Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France.
- Institut Universitaire de France, Paris, France.
- Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - John C Bell
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada.
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
25
|
Li L, Matsui Y, Prahl MK, Cassidy AG, Golan Y, Jigmeddagva U, Ozarslan N, Lin CY, Buarpung S, Gonzalez VJ, Chidboy MA, Basilio E, Lynch KL, Song D, Jegatheesan P, Rai DS, Govindaswami B, Needens J, Rincon M, Myatt L, Taha TY, Montano M, Ott M, Greene WC, Gaw SL. Neutralizing and binding antibody responses to SARS-CoV-2 with hybrid immunity in pregnancy. NPJ Vaccines 2024; 9:156. [PMID: 39191763 PMCID: PMC11349990 DOI: 10.1038/s41541-024-00948-3] [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: 04/17/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Hybrid immunity against SARS-CoV-2 has not been well studied in pregnancy. We conducted a comprehensive analysis of neutralizing antibodies (nAb) and binding antibodies in pregnant individuals who received mRNA vaccination, natural infection, or both. A third vaccine dose augmented nAb levels compared to the two-dose regimen or natural infection alone; this effect was more pronounced in hybrid immunity. There was reduced anti-Omicron nAb, but the maternal-fetal transfer efficiency remained comparable to that of other variants. Vaccine-induced nAbs were transferred more efficiently than infection-induced nAbs. Anti-spike receptor binding domain (RBD) IgG was associated with nAb against wild-type (Wuhan-Hu-1) following breakthrough infection. Both vaccination and infection-induced anti-RBD IgA, which was more durable than anti-nucleocapsid IgA. IgA response was attenuated in pregnancy compared to non-pregnant controls. These data provide additional evidence of augmentation of humoral immune responses in hybrid immunity in pregnancy.
Collapse
Affiliation(s)
- Lin Li
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yusuke Matsui
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Mary K Prahl
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Division of Pediatric Infectious Diseases and Global Health, University of California San Francisco, San Francisco, CA, USA
| | - Arianna G Cassidy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Unurzul Jigmeddagva
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Christine Y Lin
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Veronica J Gonzalez
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Megan A Chidboy
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Emilia Basilio
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Dongli Song
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Priya Jegatheesan
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, USA
| | - Daljeet S Rai
- Stanford-O'Connor Family Medicine Residency Program, Division of Family Medicine, Stanford University, Palo Alto, CA, USA
| | - Balaji Govindaswami
- Division of Neonatology, Department of Pediatrics, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Jordan Needens
- Department of Obstetrics and Gynecology, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Monica Rincon
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA.
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Departments of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA.
| | - Stephanie L Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
26
|
Wee EGT, Kempster S, Ferguson D, Hall J, Ham C, Morris S, Crook A, Gilbert SC, Korber B, Almond N, Hanke T. Design, Immunogenicity and Preclinical Efficacy of the ChAdOx1.COVconsv12 Pan-Sarbecovirus T-Cell Vaccine. Vaccines (Basel) 2024; 12:965. [PMID: 39339997 PMCID: PMC11436245 DOI: 10.3390/vaccines12090965] [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: 07/29/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
During the COVID-19 pandemic, antibody-based vaccines targeting the SARS-CoV-2 spike glycoprotein were the focus for development because neutralizing antibodies were associated with protection against the SARS-CoV-2 infection pre-clinically and in humans. While deploying these spike-based vaccines saved millions of lives worldwide, it has become clear that the immunological mechanisms of protection against severe disease are multifaceted and involve non-neutralizing antibody components. Here, we describe a novel pan-sarbecovirus T-cell vaccine, ChAdOx1.COVconsv12, designed to complement and broaden the protection of spike vaccines. The vaccine immunogen COVconsv12 employs the two regions in the viral proteome most conserved among sarbecoviruses, which are delivered by replication-deficient vector ChAdOx1. It directs T cells towards epitopes shared among sarbecoviruses including evolving SARS-CoV-2 variants. Here, we show that ChAdOx1.COVconsv12 induced broad T-cell responses in the BALB/c and C57BL/6 mice. In the Syrian hamster challenge model, ChAdOx1.COVconsv12 alone did not protect against the SARS-CoV-2 infection, but when co-administered with 1/50th of the ChAdOx1 nCoV-19 spike vaccine protective dose, faster recovery and lower oral swab viral load were observed. Induction of CD8+ T cells may decrease COVID-19 severity and extend the T-cell response coverage of variants to match the known (and as yet unknown) members of the β-coronavirus family.
Collapse
Affiliation(s)
- Edmund G.-T. Wee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
| | - Sarah Kempster
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Deborah Ferguson
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Joanna Hall
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Claire Ham
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK (S.C.G.)
| | - Alison Crook
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
| | - Sarah C. Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK (S.C.G.)
| | - Bette Korber
- New Mexico Consortium, Los Alamos, NM 87544, USA;
| | - Neil Almond
- Science and Research—Diagnostics, Medicines and Healthcare products Regulatory Agency, Potters Bar EN6 3QG, UK; (S.K.); (D.F.); (J.H.); (C.H.); (N.A.)
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK; (E.G.-T.W.); (A.C.)
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
27
|
Zhao XJ, Liu XL, Gu HJ, Liu T, Li DY, Zhang S, Wu J, Du KG, Tian S, Chen JJ, Xu Q, Lv CL, Jiang BG, Wang H, Kou ZQ, Wang GL, Fang LQ. SARS-CoV-2 reinfection broadens the antibody responses and promotes the phenotypic differentiation of virus-specific memory T cells in adolescents. J Med Virol 2024; 96:e29873. [PMID: 39165041 DOI: 10.1002/jmv.29873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/10/2024] [Accepted: 08/10/2024] [Indexed: 08/22/2024]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron subvariants raises concerns regarding the effectiveness of immunity acquired from previous Omicron subvariants breakthrough infections (BTIs) or reinfections (RIs) against the current circulating Omicron subvariants. In this study, we prospectively investigate the dynamic changes of virus-specific antibody and T cell responses among 77 adolescents following Omicron BA.2.3 BTI with or without subsequent Omicron BA.5 RI. Notably, the neutralizing antibodies (NAbs) titers against various detected SARS-CoV-2 variants, especially the emerging Omicron CH.1.1, XBB.1.5, XBB.1.16, EG.5.1, and JN.1 subvariants, exhibited a significant decrease along the time. A lower level of IgG and NAbs titers post-BTI was found to be closely associated with subsequent RI. Elevated NAbs levels and shortened antigenic distances were observed following Omicron BA.5 RI. Robust T cell responses against both Omicron BA.2- and CH.1.1-spike peptides were observed at each point visited. The exposure to Omicron BA.5 promoted phenotypic differentiation of virus-specific memory T cells, even among the non-seroconversion adolescents. Therefore, updated vaccines are needed to provide effective protection against newly emerging SARS-CoV-2 variants among adolescents.
Collapse
Affiliation(s)
- Xin-Jing Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Xiao-Lin Liu
- Institute of Infectious Disease Control and Prevention, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Hong-Jing Gu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Ti Liu
- Institute of Infectious Disease Control and Prevention, Shandong Center for Disease Control and Prevention, Jinan, China
| | - De-Yu Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Sheng Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Jie Wu
- Department of Infectious Disease Control and Prevention, Binzhou Center for Disease Control and Prevention, Jinan, China
| | - Kai-Ge Du
- Institute of Infectious Disease Control and Prevention, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Shen Tian
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Jin-Jin Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Qiang Xu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Chen-Long Lv
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Bao-Gui Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Hui Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Zeng-Qiang Kou
- Institute of Infectious Disease Control and Prevention, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Guo-Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| | - Li-Qun Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, China
| |
Collapse
|
28
|
Dallan B, Proietto D, De Laurentis M, Gallerani E, Martino M, Ghisellini S, Zurlo A, Volpato S, Govoni B, Borghesi M, Albanese V, Appay V, Bonnini S, Llewellyn-Lacey S, Pacifico S, Grumiro L, Brandolini M, Semprini S, Sambri V, Ladell K, Parry HM, Moss PAH, Price DA, Caputo A, Gavioli R, Nicoli F. Age differentially impacts adaptive immune responses induced by adenoviral versus mRNA vaccines against COVID-19. NATURE AGING 2024; 4:1121-1136. [PMID: 38918602 DOI: 10.1038/s43587-024-00644-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 05/02/2024] [Indexed: 06/27/2024]
Abstract
Adenoviral and mRNA vaccines encoding the viral spike (S) protein have been deployed globally to contain severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Older individuals are particularly vulnerable to severe infection, probably reflecting age-related changes in the immune system, which can also compromise vaccine efficacy. It is nonetheless unclear to what extent different vaccine platforms are impacted by immunosenescence. Here, we evaluated S protein-specific immune responses elicited by vaccination with two doses of BNT162b2 or ChAdOx1-S and subsequently boosted with a single dose of BNT162b2 or mRNA-1273, comparing age-stratified participants with no evidence of previous infection with SARS-CoV-2. We found that aging profoundly compromised S protein-specific IgG titers and further limited S protein-specific CD4+ and CD8+ T cell immunity as a probable function of progressive erosion of the naive lymphocyte pool in individuals vaccinated initially with BNT162b2. Our results demonstrate that primary vaccination with ChAdOx1-S and subsequent boosting with BNT162b2 or mRNA-1273 promotes sustained immunological memory in older adults and potentially confers optimal protection against coronavirus disease 2019.
Collapse
Affiliation(s)
- Beatrice Dallan
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Davide Proietto
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Martina De Laurentis
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Eleonora Gallerani
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Mara Martino
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Sara Ghisellini
- Laboratory of Clinical Pathology, University Hospital St. Anna, Ferrara, Italy
| | - Amedeo Zurlo
- Department of Medical Sciences, University of Ferrara, Geriatrics Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Stefano Volpato
- Department of Medical Sciences, University of Ferrara, Geriatrics Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Benedetta Govoni
- Department of Medical Sciences, University of Ferrara, Geriatrics Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Michela Borghesi
- Department of Economics and Management, University of Ferrara, Ferrara, Italy
| | - Valentina Albanese
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| | - Victor Appay
- Université de Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, Bordeaux, France
| | - Stefano Bonnini
- Department of Economics and Management, University of Ferrara, Ferrara, Italy
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Salvatore Pacifico
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Laura Grumiro
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Martina Brandolini
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Simona Semprini
- Unit of Microbiology, Greater Romagna Area Hub Laboratory, Cesena, Italy
| | - Vittorio Sambri
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Unit of Microbiology, Greater Romagna Area Hub Laboratory, Cesena, Italy
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Helen M Parry
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Paul A H Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, UK
| | - Antonella Caputo
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Riccardo Gavioli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco Nicoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
29
|
Zheng X, Lu R, Pan D, Peng L, He R, Hu Y, Chen J, Tang J, Rong X, Teng S, Wang Y, Liu F, Xie T, Wu C, Tang Y, Liu W, Qu X. Regulatory T and CXCR3+ Circulating Tfh Cells Concordantly Shape the Neutralizing Antibody Responses in Individuals Who Have Recovered from Mild COVID-19. J Infect Dis 2024; 230:28-37. [PMID: 39052730 DOI: 10.1093/infdis/jiae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 02/09/2024] Open
Abstract
Regulatory T (Treg) cells are involved in the antiviral immune response in patients with coronavirus disease 2019 (COVID-19); however, whether Treg cells are involved in the neutralizing antibody (nAb) response remains unclear. Here, we found that individuals who recovered from mild but not severe COVID-19 had significantly greater frequencies of Treg cells and lower frequencies of CXCR3+ circulating T follicular helper (cTfh) cells than healthy controls. Furthermore, the frequencies of Treg and CXCR3+ cTfh cells were negatively and positively correlated with the nAb responses, respectively, and Treg cells was inversely associated with CXCR3+ cTfh cells in individuals who recovered from mild COVID-19 but not in those with severe disease. Mechanistically, Treg cells inhibited memory B-cell differentiation and antibody production by limiting the activation and proliferation of cTfh cells, especially CXCR3+ cTfh cells, and functional molecule expression. This study provides novel insight showing that mild COVID-19 elicits concerted nAb responses, which are shaped by both Treg and Tfh cells.
Collapse
Affiliation(s)
- Xingyu Zheng
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Rui Lu
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Dong Pan
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Liting Peng
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Rongzhang He
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Yabin Hu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Jun Chen
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Jinyong Tang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Xiaohan Rong
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Shishan Teng
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - You Wang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
- School of Public Health, University of South China, Hengyang, China
| | - Fen Liu
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Tianyi Xie
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Chanfeng Wu
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
| | - Yinggen Tang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, Chenzhou, China
- School of Public Health, University of South China, Hengyang, China
| | - Wenpei Liu
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiaowang Qu
- College of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
30
|
Zhao XJ, Ji B, Shang C, Li DY, Zhang S, Gu HJ, Peng HH, Qian C, Zhang CL, Shi C, Shen Y, Chen JJ, Xu Q, Lv CL, Jiang BG, Wang H, Li X, Wang GL, Fang LQ. Humoral and cellular immune responses following Omicron BA.2.2 breakthrough infection and Omicron BA.5 reinfection. iScience 2024; 27:110283. [PMID: 39040063 PMCID: PMC11260851 DOI: 10.1016/j.isci.2024.110283] [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: 12/11/2023] [Revised: 04/23/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
The emergence of novel Omicron subvariants has raised concerns regarding the efficacy of immunity induced by prior Omicron subvariants breakthrough infection (BTI) or reinfection against current circulating Omicron subvariants. Here, we prospectively investigated the durability of antibody and T cell responses in individuals post Omicron BA.2.2 BTI, with or without subsequent Omicron BA.5 reinfection. Our findings reveal that the emerging Omicron subvariants, including CH.1.1, XBB, and JN.1, exhibit extensive immune evasion induced by previous infections. Notably, the level of IgG and neutralizing antibodies were found to correlate with subsequent Omicron BA.5 reinfection. Fortunately, T cell responses recognizing both Omicron BA.2 and CH.1.1 peptides were observed. Furthermore, Omicron BA.5 reinfection may alleviate immune imprinting induced by WT-vaccination, bolster virus-specific ICS+ T cell responses, and promote the phenotypic differentiation of virus-specific memory CD8+ T cells. Antigen-updated or T cell-conserved vaccines are needed to control the transmission of diverse emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Xin-Jing Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Bin Ji
- Department of Disease Control, the Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Chao Shang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - De-Yu Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Sheng Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Hong-Jing Gu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Hong-Hong Peng
- Department of Disease Control, the Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Cheng Qian
- Jiangyin Center for Disease Control and Prevention, Jiangyin, China
| | - Cui-Ling Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Chao Shi
- Department of Disease Control, the Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Yuan Shen
- Department of Disease Control, the Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Jin-Jin Chen
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Qiang Xu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Chen-Long Lv
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Bao-Gui Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Hui Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Xiao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Guo-Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| | - Li-Qun Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Science, Beijing, P.R. China
| |
Collapse
|
31
|
Qui M, Hariharaputran S, Hang SK, Zhang J, Tan CW, Chong CY, Low J, Wang L, Bertoletti A, Yung CF, Le Bert N. T cell hybrid immunity against SARS-CoV-2 in children: a longitudinal study. EBioMedicine 2024; 105:105203. [PMID: 38896919 PMCID: PMC11237860 DOI: 10.1016/j.ebiom.2024.105203] [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: 01/26/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Hybrid immunity to SARS-CoV-2, resulting from both vaccination and natural infection, remains insufficiently understood in paediatric populations, despite increasing rates of breakthrough infections among vaccinated children. METHODS We conducted a prospective longitudinal study to investigate the magnitude, specificity, and cytokine profile of antigen-specific T cell responses elicited by breakthrough SARS-CoV-2 infection in a cohort of mRNA-vaccinated children (n = 29) aged 5-11. This longitudinal analysis involved six distinct time points spanning a 16-month period post-vaccination, during which we analysed a total of 159 blood samples. All children who were followed for at least 12 months (n = 26) experienced a breakthrough infection. We conducted cytokine release assays using minimal blood samples, and we verified the cellular origin of these responses through intracellular cytokine staining. FINDINGS After breakthrough infection, children who had received mRNA vaccines showed enhanced Th1 responses specific to Spike peptides. Additionally, their Spike-specific T cells exhibited a distinctive enrichment of CD4+ IFN-γ+IL10+ cells, a characteristic akin to adults with hybrid immunity. Importantly, vaccination did not impede the development of multi-specific T cell responses targeting Membrane, Nucleoprotein, and ORF3a/7/8 antigens. INTERPRETATION Children, previously primed with a Spike-based mRNA vaccine and experiencing either symptomatic or asymptomatic breakthrough infection, retained the ability to enhance and diversify Th1/IL-10 antigen-specific T cell responses against multiple SARS-CoV-2 proteins. These findings mirror characteristics associated with hybrid cellular immunity in adults, known to confer resistance against severe COVID-19. FUNDING This study was funded by the National Medical Research Council (NMRC) Singapore (COVID19RF-0019, MOH-000019, MOH-000535, OFLCG19May-0034 and MOH-OFYIRG19nov-0002).
Collapse
Affiliation(s)
- Martin Qui
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Shou Kit Hang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Jinyan Zhang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chia Yin Chong
- KK Women's and Children's Hospital, Department of Paediatrics, Infectious Diseases Service, Singapore; Duke-NUS Medical School, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jenny Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Singapore General Hospital, Department of Infectious Diseases, Singapore
| | - Linfa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Singapore Immunology Network, A∗STAR, Singapore
| | - Chee Fu Yung
- KK Women's and Children's Hospital, Department of Paediatrics, Infectious Diseases Service, Singapore; Duke-NUS Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.
| |
Collapse
|
32
|
Hernandez-Galicia G, Gomez-Morales L, Lopez-Bailon LU, Valdovinos-Torres H, Contreras-Ochoa CO, Díaz Benítez CE, Martinez-Barnetche J, Alpuche-Aranda C, Ortiz-Navarrete V. Presence of SARS-CoV-2-specific T cells before vaccination in the Mexican population. J Leukoc Biol 2024; 116:95-102. [PMID: 38717738 DOI: 10.1093/jleuko/qiae111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 06/30/2024] Open
Abstract
The immune response to SARS-CoV-2 has been extensively studied following the pandemic outbreak in 2020; however, the presence of specific T cells against SARS-CoV-2 before vaccination has not been evaluated in Mexico. In this study, we estimated the frequency of T CD4+ and T CD8+ cells that exhibit a specific response to S (spike) and N (nucleocapsid) proteins in a Mexican population. We collected 78 peripheral blood samples from unvaccinated subjects, and the presence of antibodies against spike (RBD) and N protein was determined. Peripheral blood mononuclear cells were isolated and stimulated with a pool of S or N protein peptides (Wuhan-Hu-1 strain). IL-1β, IL-4, IL-6, IL-10, IL-2, IL-8, TNF-α, IFN-γ, and GM-CSF levels were quantified in the supernatant of the activated cells, and the cells were stained to assess the activation and memory phenotypes. Differential activation frequency dependent on serological status was observed in CD4+ cells but not in CD8+ cells. The predominantly activated population was the central memory T CD4+ cells. Only 10% of the population exhibited the same phenotype with respect to the response to nucleocapsid peptides. The cytokine profile differed between the S and N responses. S peptides induced a more proinflammatory response compared with the N peptides. In conclusion, in a Mexican cohort before vaccination, there was a significant response to the S and N SARS-CoV-2 proteins resulting from previous infections with seasonal coronaviruses or previous undetected exposure to SARS-CoV-2.
Collapse
Affiliation(s)
- Gabriela Hernandez-Galicia
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
| | - Luis Gomez-Morales
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
- Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Manuel Carpio and Plan de Ayala St, 11340, Mexico City, Mexico
| | - Luis Uriel Lopez-Bailon
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
- Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Manuel Carpio and Plan de Ayala St, 11340, Mexico City, Mexico
| | - Humberto Valdovinos-Torres
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Carla O Contreras-Ochoa
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Cinthya Estefhany Díaz Benítez
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Jesus Martinez-Barnetche
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Celia Alpuche-Aranda
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
| |
Collapse
|
33
|
Fan X, Song JW, Cao WJ, Zhou MJ, Yang T, Wang J, Meng FP, Shi M, Zhang C, Wang FS. T-Cell Epitope Mapping of SARS-CoV-2 Reveals Coordinated IFN-γ Production and Clonal Expansion of T Cells Facilitates Recovery from COVID-19. Viruses 2024; 16:1006. [PMID: 39066169 PMCID: PMC11281491 DOI: 10.3390/v16071006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/01/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND T-cell responses can be protective or detrimental during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection; however, the underlying mechanism is poorly understood. METHODS In this study, we screened 144 15-mer peptides spanning the SARS-CoV-2 spike, nucleocapsid (NP), M, ORF8, ORF10, and ORF3a proteins and 39 reported SARS-CoV-1 peptides in peripheral blood mononuclear cells (PBMCs) from nine laboratory-confirmed coronavirus disease 2019 (COVID-19) patients (five moderate and four severe cases) and nine healthy donors (HDs) collected before the COVID-19 pandemic. T-cell responses were monitored by IFN-γ and IL-17A production using ELISA, and the positive samples were sequenced for the T cell receptor (TCR) β chain. The positive T-cell responses to individual SARS-CoV-2 peptides were validated by flow cytometry. RESULTS COVID-19 patients with moderate disease produced more IFN-γ than HDs and patients with severe disease (moderate vs. HDs, p < 0.0001; moderate vs. severe, p < 0.0001) but less IL-17A than those with severe disease (p < 0.0001). A positive correlation was observed between IFN-γ production and T-cell clonal expansion in patients with moderate COVID-19 (r = 0.3370, p = 0.0214) but not in those with severe COVID-19 (r = -0.1700, p = 0.2480). Using flow cytometry, we identified that a conserved peptide of the M protein (Peptide-120, P120) was a dominant epitope recognized by CD8+ T cells in patients with moderate disease. CONCLUSION Coordinated IFN-γ production and clonal expansion of SARS-CoV-2-specific T cells are associated with disease resolution in COVID-19. Our findings contribute to a better understanding of T-cell-mediated immunity in COVID-19 and may inform future strategies for managing and preventing severe outcomes of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xing Fan
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Jin-Wen Song
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wen-Jing Cao
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ming-Ju Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Tao Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Jing Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Fan-Ping Meng
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Ming Shi
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Chao Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- Medical School of Chinese PLA, Beijing 100853, China
| |
Collapse
|
34
|
Gray-Gaillard SL, Solis SM, Chen HM, Monteiro C, Ciabattoni G, Samanovic MI, Cornelius AR, Williams T, Geesey E, Rodriguez M, Ortigoza MB, Ivanova EN, Koralov SB, Mulligan MJ, Herati RS. SARS-CoV-2 inflammation durably imprints memory CD4 T cells. Sci Immunol 2024; 9:eadj8526. [PMID: 38905326 PMCID: PMC11824880 DOI: 10.1126/sciimmunol.adj8526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 05/30/2024] [Indexed: 06/23/2024]
Abstract
Memory CD4 T cells are critical to human immunity, yet it is unclear whether viral inflammation during memory formation has long-term consequences. Here, we compared transcriptional and epigenetic landscapes of Spike (S)-specific memory CD4 T cells in 24 individuals whose first exposure to S was via SARS-CoV-2 infection or mRNA vaccination. Nearly 2 years after memory formation, S-specific CD4 T cells established by infection remained enriched for transcripts related to cytotoxicity and for interferon-stimulated genes, likely because of a chromatin accessibility landscape altered by inflammation. Moreover, S-specific CD4 T cells primed by infection had reduced proliferative capacity in vitro relative to vaccine-primed cells. Furthermore, the transcriptional state of S-specific memory CD4 T cells was minimally altered by booster immunization and/or breakthrough infection. Thus, infection-associated inflammation durably imprints CD4 T cell memory, which affects the function of these cells and may have consequences for long-term immunity.
Collapse
Affiliation(s)
| | - Sabrina M. Solis
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Han M. Chen
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Clarice Monteiro
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Grace Ciabattoni
- Department of Microbiology, New York University School of
Medicine; New York, NY, USA
| | - Marie I. Samanovic
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Amber R. Cornelius
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Tijaana Williams
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Emilie Geesey
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Miguel Rodriguez
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Mila Brum Ortigoza
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Ellie N. Ivanova
- Department of Pathology, New York University School of
Medicine; New York, NY, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University School of
Medicine; New York, NY, USA
| | - Mark J. Mulligan
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
- Department of Microbiology, New York University School of
Medicine; New York, NY, USA
| | - Ramin Sedaghat Herati
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
- Department of Microbiology, New York University School of
Medicine; New York, NY, USA
| |
Collapse
|
35
|
Tarke A, Ramezani-Rad P, Alves Pereira Neto T, Lee Y, Silva-Moraes V, Goodwin B, Bloom N, Siddiqui L, Avalos L, Frazier A, Zhang Z, da Silva Antunes R, Dan J, Crotty S, Grifoni A, Sette A. SARS-CoV-2 breakthrough infections enhance T cell response magnitude, breadth, and epitope repertoire. Cell Rep Med 2024; 5:101583. [PMID: 38781962 PMCID: PMC11228552 DOI: 10.1016/j.xcrm.2024.101583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/22/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Little is known about the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 or SARS2) vaccine breakthrough infections (BTIs) on the magnitude and breadth of the T cell repertoire after exposure to different variants. We studied samples from individuals who experienced symptomatic BTIs during Delta or Omicron waves. In the pre-BTI samples, 30% of the donors exhibited substantial immune memory against non-S (spike) SARS2 antigens, consistent with previous undiagnosed asymptomatic SARS2 infections. Following symptomatic BTI, we observed (1) enhanced S-specific CD4 and CD8 T cell responses in donors without previous asymptomatic infection, (2) expansion of CD4 and CD8 T cell responses to non-S targets (M, N, and nsps) independent of SARS2 variant, and (3) generation of novel epitopes recognizing variant-specific mutations. These variant-specific T cell responses accounted for 9%-15% of the total epitope repertoire. Overall, BTIs boost vaccine-induced immune responses by increasing the magnitude and by broadening the repertoire of T cell antigens and epitopes recognized.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Parham Ramezani-Rad
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Yeji Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Vanessa Silva-Moraes
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Benjamin Goodwin
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Nathaniel Bloom
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Leila Siddiqui
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Liliana Avalos
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Zeli Zhang
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Jennifer Dan
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
36
|
Ruta S, Popescu CP, Matei L, Grancea C, Paun AM, Oprea C, Sultana C. SARS-CoV-2 Humoral and Cellular Immune Responses in People Living with HIV. Vaccines (Basel) 2024; 12:663. [PMID: 38932392 PMCID: PMC11209143 DOI: 10.3390/vaccines12060663] [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: 05/10/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Immunosuppressed individuals, such as people living with HIV (PLWH), remain vulnerable to severe COVID-19. We analyzed the persistence of specific SARS-CoV-2 humoral and cellular immune responses in a retrospective, cross-sectional study in PLWH on antiretroviral therapy. Among 104 participants, 70.2% had anti-S IgG antibodies, and 55.8% had significant neutralizing activity against the Omicron variant in a surrogate virus neutralization test. Only 38.5% were vaccinated (8.76 ± 4.1 months prior), all displaying anti-S IgG, 75% with neutralizing antibodies and anti-S IgA. Overall, 29.8% of PLWH had no SARS-CoV-2 serologic markers; they displayed significantly lower CD4 counts and higher HIV viral load. Severe immunosuppression (present in 12.5% of participants) was linked to lower levels of detectable anti-S IgG (p = 0.0003), anti-S IgA (p < 0.0001) and lack of neutralizing activity against the Omicron variant (p < 0.0001). T-cell responses were present in 86.7% of tested participants, even in those lacking serological markers. In PLWH without severe immunosuppression, neutralizing antibodies and T-cell responses persisted for up to 9 months post-infection or vaccination. Advanced immunosuppression led to diminished humoral immune responses but retained specific cellular immunity.
Collapse
Affiliation(s)
- Simona Ruta
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.R.); (C.O.); (C.S.)
- Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.M.); (C.G.)
| | - Corneliu Petru Popescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.R.); (C.O.); (C.S.)
- Dr. Victor Babes Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania;
| | - Lilia Matei
- Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.M.); (C.G.)
| | - Camelia Grancea
- Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.M.); (C.G.)
| | - Adrian Marius Paun
- Dr. Victor Babes Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania;
| | - Cristiana Oprea
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.R.); (C.O.); (C.S.)
- Dr. Victor Babes Hospital of Infectious and Tropical Diseases, 030303 Bucharest, Romania;
| | - Camelia Sultana
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.R.); (C.O.); (C.S.)
- Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.M.); (C.G.)
| |
Collapse
|
37
|
Yang G, Cao J, Qin J, Mei X, Deng S, Xia Y, Zhao J, Wang J, Luan T, Chen D, Huang P, Chen C, Sun X, Luo Q, Su J, Zhang Y, Zhong N, Wang Z. Initial COVID-19 severity influenced by SARS-CoV-2-specific T cells imprints T-cell memory and inversely affects reinfection. Signal Transduct Target Ther 2024; 9:141. [PMID: 38811527 PMCID: PMC11136975 DOI: 10.1038/s41392-024-01867-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024] Open
Abstract
The immunoprotective components control COVID-19 disease severity, as well as long-term adaptive immunity maintenance and subsequent reinfection risk discrepancies across initial COVID-19 severity, remain unclarified. Here, we longitudinally analyzed SARS-CoV-2-specific immune effectors during the acute infection and convalescent phases of 165 patients with COVID-19 categorized by severity. We found that early and robust SARS-CoV-2-specific CD4+ and CD8+ T cell responses ameliorate disease progression and shortened hospital stay, while delayed and attenuated virus-specific CD8+ T cell responses are prominent severe COVID-19 features. Delayed antiviral antibody generation rather than titer level associates with severe outcomes. Conversely, initial COVID-19 severity imprints the long-term maintenance of SARS-CoV-2-specific adaptive immunity, demonstrating that severe convalescents exhibited more sustained virus-specific antibodies and memory T cell responses compared to mild/moderate counterparts. Moreover, initial COVID-19 severity inversely correlates with SARS-CoV-2 reinfection risk. Overall, our study unravels the complicated interaction between temporal characteristics of virus-specific T cell responses and COVID-19 severity to guide future SARS-CoV-2 wave management.
Collapse
Affiliation(s)
- Gang Yang
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Jinpeng Cao
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jian Qin
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Xinyue Mei
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Shidong Deng
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yingjiao Xia
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Jun Zhao
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Junxiang Wang
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Tao Luan
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Daxiang Chen
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Peiyu Huang
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Cheng Chen
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China
| | - Xi Sun
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Qi Luo
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jie Su
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yunhui Zhang
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China.
| | - Nanshan Zhong
- The Affiliated Hospital of Kunming University of Science and Technology. Department of Respiratory and Critical Care Medicine, The First People's Hospital of Yunnan province, Kunming, Yunnan, China.
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China.
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Zhongfang Wang
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China.
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Li J, Hsu KS, Howe SE, Hoang T, Xia Z, Berzofsky JA, Sui Y. Sex-biased immunogenicity of a mucosal subunit vaccine against SARS-CoV-2 in mice. Front Immunol 2024; 15:1386243. [PMID: 38835757 PMCID: PMC11148259 DOI: 10.3389/fimmu.2024.1386243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction Current vaccines against COVID-19 administered via parenteral route have limited ability to induce mucosal immunity. There is a need for an effective mucosal vaccine to combat SARS-CoV-2 virus replication in the respiratory mucosa. Moreover, sex differences are known to affect systemic antibody responses against vaccines. However, their role in mucosal cellular responses against a vaccine remains unclear and is underappreciated. Methods We evaluated the mucosal immunogenicity of a booster vaccine regimen that is recombinant protein-based and administered intranasally in mice to explore sex differences in mucosal humoral and cellular responses. Results Our results showed that vaccinated mice elicited strong systemic antibody (Ab), nasal, and bronchiole alveolar lavage (BAL) IgA responses, and local T cell immune responses in the lung in a sex-biased manner irrespective of mouse genetic background. Monocytes, alveolar macrophages, and CD103+ resident dendritic cells (DCs) in the lungs are correlated with robust mucosal Ab and T cell responses induced by the mucosal vaccine. Discussion Our findings provide novel insights into optimizing next-generation booster vaccines against SARS-CoV-2 by inducing spike-specific lung T cell responses, as well as optimizing mucosal immunity for other respiratory infections, and a rationale for considering sex differences in future vaccine research and vaccination practice.
Collapse
MESH Headings
- Animals
- Female
- Mice
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19/virology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Male
- Immunity, Mucosal
- Immunogenicity, Vaccine
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Lung/immunology
- Lung/virology
- T-Lymphocytes/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Mice, Inbred C57BL
- Administration, Intranasal
- Sex Factors
- Immunoglobulin A/immunology
- Dendritic Cells/immunology
- Immunization, Secondary
- Immunity, Humoral
Collapse
Affiliation(s)
- Jianping Li
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Kevin S Hsu
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Savannah E Howe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Tanya Hoang
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Zheng Xia
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
39
|
Suthar MS. Durability of immune responses to SARS-CoV-2 infection and vaccination. Semin Immunol 2024; 73:101884. [PMID: 38861769 PMCID: PMC11490408 DOI: 10.1016/j.smim.2024.101884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024]
Abstract
Infection with SARS-CoV-2 in humans has caused a pandemic of unprecedented dimensions. SARS-CoV-2 is primarily transmitted through respiratory droplets and targets ciliated epithelial cells in the nasal cavity, trachea, and lungs by utilizing the cellular receptor angiotensin-converting enzyme 2 (ACE2). The innate immune response, including type I and III interferons, inflammatory cytokines (IL-6, TNF-α, IL-1β), innate immune cells (monocytes, DCs, neutrophils, natural killer cells), antibodies (IgG, sIgA, neutralizing antibodies), and adaptive immune cells (B cells, CD8+ and CD4+ T cells) play pivotal roles in mitigating COVID-19 disease. Broad and durable B-cell- and T-cell immunity elicited by infection and vaccination is essential for protection against severe disease, hospitalization and death. However, the emergence of SARS-CoV-2 variants that evade neutralizing antibodies continue to jeopardize vaccine efficacy. In this review, we highlight our understanding the infection- and vaccine-mediated humoral, B and T cell responses, the durability of the immune responses, and how variants continue to threaten the efficacy of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Emory Vaccine Center, Emory University, Atlanta, GA, USA; Emory Center of Excellence of Influenza Research and Response (CEIRR), Atlanta, GA, USA; Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
40
|
Yang M, Meng Y, Hao W, Zhang J, Liu J, Wu L, Lin B, Liu Y, Zhang Y, Yu X, Wang X, Gong Y, Ge L, Fan Y, Xie C, Xu Y, Chang Q, Zhang Y, Qin X. A prognostic model for SARS-CoV-2 breakthrough infection: Analyzing a prospective cellular immunity cohort. Int Immunopharmacol 2024; 131:111829. [PMID: 38489974 DOI: 10.1016/j.intimp.2024.111829] [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: 12/08/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Following the COVID-19 pandemic, studies have identified several prevalent characteristics, especially related to lymphocyte subsets. However, limited research is available on the focus of this study, namely, the specific memory cell subsets among individuals who received COVID-19 vaccine boosters and subsequently experienced a SARS-CoV-2 breakthrough infection. METHODS Flow cytometry (FCM) was employed to investigate the early and longitudinal pattern changes of cellular immunity in patients with SARS-CoV-2 breakthrough infections following COVID-19 vaccine boosters. XGBoost (a machine learning algorithm) was employed to analyze cellular immunity prior to SARS-CoV-2 breakthrough, aiming to establish a prognostic model for SARS-CoV-2 breakthrough infections. RESULTS Following SARS-CoV-2 breakthrough infection, naïve T cells and TEMRA subsets increased while the percentage of TCM and TEM cells decreased. Naïve and non-switched memory B cells increased while switched and double-negative memory B cells decreased. The XGBoost model achieved an area under the curve (AUC) of 0.78, with an accuracy rate of 81.8 %, a sensitivity of 75 %, and specificity of 85.7 %. TNF-α, CD27-CD19+cells, and TEMRA subsets were identified as high predictors. An increase in TNF-α, cTfh, double-negative memory B cells, IL-6, IL-10, and IFN-γ prior to SARS-CoV-2 infection was associated with enduring clinical symptoms; conversely, an increase in CD3+ T cells, CD4+ T cells, and IL-2 was associated with clinical with non-enduring clinical symptoms. CONCLUSION SARS-CoV-2 breakthrough infection leads to disturbances in cellular immunity. Assessing cellular immunity prior to breakthrough infection serves as a valuable prognostic tool for SARS-CoV-2 infection, which facilitates clinical decision-making.
Collapse
Affiliation(s)
- Mei Yang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yuan Meng
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Wudi Hao
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jin Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jianhua Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Baoxu Lin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yong Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yue Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaojun Yu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaoqian Wang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yu Gong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lili Ge
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yan Fan
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Conghong Xie
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yiyun Xu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qing Chang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yixiao Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Xiaosong Qin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
41
|
Chen Y, Zhao T, Chen L, Jiang G, Geng Y, Li W, Yin S, Tong X, Tao Y, Ni J, Lu Q, Ning M, Wu C. SARS-CoV-2 Omicron infection augments the magnitude and durability of systemic and mucosal immunity in triple-dose CoronaVac recipients. mBio 2024; 15:e0240723. [PMID: 38456703 PMCID: PMC11005357 DOI: 10.1128/mbio.02407-23] [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: 09/06/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024] Open
Abstract
The inactivated whole-virion vaccine, CoronaVac, is one of the most widely used coronavirus disease 2019 (COVID-19) vaccines worldwide. There is a paucity of data indicating the durability of the immune response and the impact of immune imprinting induced by CoronaVac upon Omicron infection. In this prospective cohort study, 41 recipients of triple-dose CoronaVac and 14 unvaccinated individuals were recruited. We comprehensively profiled adaptive immune parameters in both groups, including spike-specific immunoglobulin (Ig) G and IgA titers, neutralizing activity, B cells, circulating follicular helper T (cTfh) cells, CD4+ and CD8+ T cells, and their memory subpopulations at 12 months after the third booster dose and at 4 and 20 weeks after Omicron BA.5 infection. Twelve months after the third CoronaVac vaccination, spike-specific antibodies and cellular responses were detectable in most vaccinated individuals. BA.5 infection significantly augmented the magnitude, cross-reactivity, and durability of serum neutralization activities, Fc-mediated phagocytosis, nasal spike-specific IgA responses, memory B cells, activated cTfh cells, memory CD4+ T cells, and memory CD8+ T cells for both the ancestral strain and Omicron subvariants, compared to unvaccinated individuals. Notably, the increase in BA.5-specific immunity after breakthrough infection was consistently comparable to or higher than that of the ancestral strain, suggesting no evidence of immune imprinting. Immune landscape analyses showed that vaccinated individuals have better synchronization of multiple immune components than unvaccinated individuals upon heterologous infection. Our data provide detailed insight into the protective role of the inactivated COVID-19 vaccine in shaping humoral and cellular immunity to Omicron infection. IMPORTANCE There is a paucity of data indicating the durability of the immune response and the impact of immune imprinting induced by CoronaVac upon Omicron breakthrough infection. In this prospective cohort study, the anti-severe acute respiratory syndrome coronavirus 2 adaptive responses were analyzed before and after the Omicron BA.5 infection. Our data provide detailed insight into the protective role of the inactivated COVID-19 vaccine in shaping humoral and cellular immune responses to heterologous Omicron infection. CLINICAL TRIAL This study is registered with ClinicalTrials.gov as NCT05680896.
Collapse
Affiliation(s)
- Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Tiantian Zhao
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Geng
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wanting Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - Shengxia Yin
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xin Tong
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Yue Tao
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Ni
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qiuhan Lu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mingzhe Ning
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Chao Wu
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
42
|
Owens K, Esmaeili S, Schiffer JT. Heterogeneous SARS-CoV-2 kinetics due to variable timing and intensity of immune responses. JCI Insight 2024; 9:e176286. [PMID: 38573774 PMCID: PMC11141931 DOI: 10.1172/jci.insight.176286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
The viral kinetics of documented SARS-CoV-2 infections exhibit a high degree of interindividual variability. We identified 6 distinct viral shedding patterns, which differed according to peak viral load, duration, expansion rate, and clearance rate, by clustering data from 768 infections in the National Basketball Association cohort. Omicron variant infections in previously vaccinated individuals generally led to lower cumulative shedding levels of SARS-CoV-2 than other scenarios. We then developed a mechanistic mathematical model that recapitulated 1,510 observed viral trajectories, including viral rebound and cases of reinfection. Lower peak viral loads were explained by a more rapid and sustained transition of susceptible cells to a refractory state during infection as well as by an earlier and more potent late, cytolytic immune response. Our results suggest that viral elimination occurs more rapidly during Omicron infection, following vaccination, and following reinfection due to enhanced innate and acquired immune responses. Because viral load has been linked with COVID-19 severity and transmission risk, our model provides a framework for understanding the wide range of observed SARS-CoV-2 infection outcomes.
Collapse
Affiliation(s)
- Katherine Owens
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shadisadat Esmaeili
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Joshua T. Schiffer
- Fred Hutchinson Cancer Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
- University of Washington, Department of Medicine, Seattle, Washington, USA
| |
Collapse
|
43
|
Bagnoli F, Galgani I, Vadivelu VK, Phogat S. Reverse development of vaccines against antimicrobial-resistant pathogens. NPJ Vaccines 2024; 9:71. [PMID: 38570502 PMCID: PMC10991305 DOI: 10.1038/s41541-024-00858-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Vaccine R&D is typically a lengthy process taking >10 years. However, vaccines still fail in clinical development because of unreliable animal models or absent immunological correlates of protection. Without a correlate of protection, phase-1 and -2 studies of safety and immunogenicity can fail to predict phase-3 efficacy. Indeed, the history of vaccine development is replete with promising phase-1 and -2 results and failed phase-3 efficacy trials. To avoid this misfortune, we present Reverse Vaccine Development for vaccines against antimicrobial-resistant (AMR) pathogens. In this approach, instead of evaluating efficacy in phase 3, proof-of-principle efficacy is evaluated as early as possible in a population with a high incidence of disease, which may differ from the population intended for registration, and can be a controlled human infection population. To identify a correlate of protection in these populations, the vaccine-elicited immune response is compared between protected and unprotected subjects. If a correlate is identified, it can help to refine the vaccine dosage, schedule, and formulation, and facilitate the assessment of vaccine efficacy in other populations with different attack rates, subject characteristics, and disease manifestations. This may be the only way to provide life-saving vaccines to populations affected by AMR-pathogen diseases at incidences that are typically low and unsuited to phase-3 efficacy trials. The availability of a correlate of protection early in clinical development can potentially prevent failures of large phase-3 trials and unnecessary exposures of populations to inefficacious vaccines that have resulted in disinvestment in the development of vaccines against AMR pathogens.
Collapse
|
44
|
Müller TR, Buggert M. Boosting SARS-CoV-2 immunity in immunocompromised individuals. Genes Immun 2024; 25:168-169. [PMID: 38114640 PMCID: PMC11023933 DOI: 10.1038/s41435-023-00219-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 12/21/2023]
Affiliation(s)
- Thomas R Müller
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
45
|
Sohail MS, Ahmed SF, Quadeer AA, McKay MR. Cross-Reactivity Assessment of Vaccine-Derived SARS-CoV-2 T Cell Responses against BA.2.86 and JN.1. Viruses 2024; 16:473. [PMID: 38543838 PMCID: PMC10975570 DOI: 10.3390/v16030473] [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: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 05/23/2024] Open
Abstract
The SARS-CoV-2 Omicron sub-variants BA.2.86 and JN.1 contain multiple mutations in the spike protein that were not present in previous variants of concern and Omicron sub-variants. Preliminary research suggests that these variants reduce the neutralizing capability of antibodies induced by vaccines, which is particularly significant for JN.1. This raises concern as many widely deployed COVID-19 vaccines are based on the spike protein of the ancestral Wuhan strain of SARS-CoV-2. While T cell responses have been shown to be robust against previous SARS-CoV-2 variants, less is known about the impact of mutations in BA.2.86 and JN.1 on T cell responses. We evaluate the effect of mutations specific to BA.2.86 and JN.1 on experimentally determined T cell epitopes derived from the spike protein of the ancestral Wuhan strain and the spike protein of the XBB.1.5 strain that has been recommended as a booster vaccine. Our data suggest that BA.2.86 and JN.1 affect numerous T cell epitopes in spike compared to previous variants; however, the widespread loss of T cell recognition against these variants is unlikely.
Collapse
Affiliation(s)
- Muhammad Saqib Sohail
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong SAR, China;
| | - Syed Faraz Ahmed
- Department of Electrical and Electronic Engineering, University of Melbourne, Parkville, VIC 3010, Australia;
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Ahmed Abdul Quadeer
- Department of Electrical and Electronic Engineering, University of Melbourne, Parkville, VIC 3010, Australia;
| | - Matthew R. McKay
- Department of Electrical and Electronic Engineering, University of Melbourne, Parkville, VIC 3010, Australia;
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
46
|
Choi MJ, Hyun H, Heo JY, Seo YB, Noh JY, Cheong HJ, Kim WJ, Kim HJ, Choi JY, Lee YJ, Chung EJ, Kim SH, Jeong H, Kim B, Song JY. Longitudinal immune kinetics of COVID-19 booster versus primary series vaccination: Insight into the annual vaccination strategy. Heliyon 2024; 10:e27211. [PMID: 38468934 PMCID: PMC10926122 DOI: 10.1016/j.heliyon.2024.e27211] [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: 08/25/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
Background Data on the durability of booster dose immunity of COVID-19 vaccines are relatively limited. Methods Immunogenicity was evaluated for up to 9-12 months after the third dose of vaccination in 94 healthy adults. Results Following the third dose, the anti-spike immunoglobulin G (IgG) antibody response against the wild-type was boosted markedly, which decreased gradually over time. However, even 9-12 months after the booster dose, both the median and geometric mean of anti-spike IgG antibody levels were higher than those measured 4 weeks after the second dose. Breakthrough infection during the Omicron-dominant period boosted neutralizing antibody titers against Omicron sublineages (BA.1 and BA.5) and the ancestral strain. T-cell immune response was efficiently induced and maintained during the study period. Conclusions mRNA vaccine booster dose elicited durable humoral immunity for up to 1 year after the third dose and T-cell immunity was sustained during the study period, supporting an annual COVID-19 vaccination strategy.
Collapse
Affiliation(s)
- Min Joo Choi
- Department of Internal Medicine, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, Republic of Korea
| | - Hakjun Hyun
- Department of Infectious Diseases, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jung Yeon Heo
- Department of Infectious Diseases, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yu Bin Seo
- Division of Infectious Disease, Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Ji Yun Noh
- Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Vaccine Innovation Center - Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Vaccine Innovation Center - Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo Joo Kim
- Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Vaccine Innovation Center - Korea University College of Medicine, Seoul, Republic of Korea
| | - Hwa Jung Kim
- Department of Clinical Epidemiology and Biostatistics, ASAN Medical Center, Ulsan University College of Medicine, Seoul, Republic of Korea
| | - Ju-yeon Choi
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, Cheongju, Republic of Korea
| | - Young Jae Lee
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, Cheongju, Republic of Korea
| | - Eun Joo Chung
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, Cheongju, Republic of Korea
| | - Su-Hwan Kim
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, Cheongju, Republic of Korea
| | - Hyeonji Jeong
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, Cheongju, Republic of Korea
| | - Byoungguk Kim
- Division of Vaccine Clinical Research, Center for Vaccine Research, National Institute of Infectious Diseases, Cheongju, Republic of Korea
| | - Joon Young Song
- Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
- Vaccine Innovation Center - Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
47
|
Perera DJ, Koger-Pease C, Paulini K, Daoudi M, Ndao M. Beyond schistosomiasis: unraveling co-infections and altered immunity. Clin Microbiol Rev 2024; 37:e0009823. [PMID: 38319102 PMCID: PMC10938899 DOI: 10.1128/cmr.00098-23] [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] [Indexed: 02/07/2024] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kayla Paulini
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Mohamed Daoudi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
48
|
Sanchez S, Dangi T, Awakoaiye B, Irani N, Fourati S, Richner J, Penaloza-MacMaster P. Time-dependent enhancement of mRNA vaccines by 4-1BB costimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582992. [PMID: 38496467 PMCID: PMC10942304 DOI: 10.1101/2024.03.01.582992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
mRNA vaccines have demonstrated efficacy against COVID-19. However, concerns regarding waning immunity and breakthrough infections have motivated the development of next-generation vaccines with enhanced efficacy. In this study, we investigated the impact of 4-1BB costimulation on immune responses elicited by mRNA vaccines in mice. We first vaccinated mice with an mRNA vaccine encoding the SARS-CoV-2 spike antigen like the Moderna and Pfizer-BioNTech vaccines, followed by administration of 4-1BB costimulatory antibodies at various times post-vaccination. Administering 4-1BB costimulatory antibodies during the priming phase did not enhance immune responses. However, administering 4-1BB costimulatory antibodies after 96 hours elicited a significant improvement in CD8 T cell responses, leading to enhanced protection against breakthrough infections. A similar improvement in immune responses was observed with multiple mRNA vaccines, including vaccines against common cold coronavirus, human immunodeficiency virus (HIV), and arenavirus. These findings demonstrate a time-dependent effect by 4-1BB costimulation and provide insights for developing improved mRNA vaccines.
Collapse
Affiliation(s)
- Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Bakare Awakoaiye
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nahid Irani
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Slim Fourati
- Department of Medicine, Division of Allergy and Immunology, Feinberg School of Medicine and Center for Human Immunobiology, Northwestern University, Chicago, IL 60611, USA
| | - Justin Richner
- Department of Microbiology & Immunology, University of Illinois Chicago College of Medicine, Chicago, IL 60612, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
49
|
Zha G, Chen Z, Wu N, Huang T, Deng Z, Cai D, Peng M, Hu P, Ren H. Clinical characteristics and immunogenicity after Omicron breakthrough infection in patients with chronic hepatitis B infection: A longitudinal observational study. J Med Virol 2024; 96:e29548. [PMID: 38511555 DOI: 10.1002/jmv.29548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
The clinical and immunological features after breakthrough infection (BTI) during Omicron wave in patients with chronic hepatitis B virus infection (CHB) are still unclear. A total of 101 patients with CHB from our previous coronavirus disease 2019 (COVID-19) vaccination cohort (NCT05007665), were continued to be followed up at the Second Affiliated Hospital of Chongqing Medical University after BTI, while an additional 39 healthcare workers after BTI were recruited as healthy controls (HCs). Clinical data were collected using questionnaire survey and electronic medical record. Blood samples were used to determine the antibody responses, as well as B and T cell responses. After BTI, the clinical symptoms of COVID-19 were mild to moderate in patients with CHB, with a median duration of 5 days. Compared with HCs, patients with CHB were more susceptible to develop moderate COVID-19. The liver function was not significantly damaged, and HBV-DNA was not activated in patients with CHB after BTI. Patients with CHB could elicit robust antibody responses after BTI (NAbs 13.0-fold, BA.5 IgG: 24.2-fold, respectively), which was also significantly higher than that in every period after vaccination (all p < 0.001), and compared to that in HCs after BTI. The CD4+, cTfh, and CD8+ T cell responses were also augmented in patients with CHB after BTI, while exhibiting comparability to those observed in HCs. In patients with CHB after BTI, the immune imprint was observed in B cell responses, rather than in T cell responses. In conclusion, Omicron breakthrough infection induced mild to moderate COVID-19 symptoms in patients with CHB, without exacerbating the progress of liver diseases. Meanwhile, BTI demonstrated the ability to induce robust antibody and T cell responses in patients with CHB, which was comparable to those observed in HCs.
Collapse
Affiliation(s)
- Guanhua Zha
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiwei Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Na Wu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tianquan Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiling Deng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dachuan Cai
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingli Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Ren
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
50
|
Miyamoto S, Suzuki T. Infection-mediated immune response in SARS-CoV-2 breakthrough infection and implications for next-generation COVID-19 vaccine development. Vaccine 2024; 42:1401-1406. [PMID: 38310015 DOI: 10.1016/j.vaccine.2024.01.088] [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: 06/26/2023] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
Post-vaccination infections, termed breakthrough infections, occur after the virus infection overcomes the vaccine-induced immune barrier. During the early stages of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron wave, high serum-neutralizing antibody titers against the Omicron variant were detected in individuals with breakthrough infections as well as those who received a third vaccine dose (i.e., booster recipients). Additionally, these cases indicated that Omicron antigens triggered an immune response that differed from that triggered by the vaccine strain before analysis of the effectiveness of new vaccines updated for the Omicron variants. Moreover, the magnitude and breadth of neutralizing antibody titers induced by breakthrough infections are correlated with the upper respiratory viral load at diagnosis and the duration between vaccination and infection, respectively. Unlike booster vaccine recipients, patients with breakthrough infections have varying durations between vaccination and infection. Accordingly, optimal booster vaccination intervals may be estimated based on the cross-neutralizing antibody response induced over time. Examination of breakthrough infection cases has provided valuable insights that could not be yielded by only examining vaccinated individuals alone. These insights include estimates of vaccine-induced immunity against SARS-CoV-2 variants and the various factors related to the clinical status. This review describes the immune response elicited by breakthrough infections; specifically, it discusses factors that affect the magnitude and breadth of serum antibody titers as well as the appropriate booster vaccination strategy. This review provides key aspects that could contribute to developing next-generation COVID-19 vaccines through breakthrough infection cases.
Collapse
Affiliation(s)
- Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases Tokyo 162-8640, Japan.
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases Tokyo 162-8640, Japan
| |
Collapse
|