1
|
Yang Y, Miller H, Byazrova MG, Cndotti F, Benlagha K, Camara NOS, Shi J, Forsman H, Lee P, Yang L, Filatov A, Zhai Z, Liu C. The characterization of CD8 + T-cell responses in COVID-19. Emerg Microbes Infect 2024; 13:2287118. [PMID: 37990907 PMCID: PMC10786432 DOI: 10.1080/22221751.2023.2287118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/19/2023] [Indexed: 11/23/2023]
Abstract
This review gives an overview of the protective role of CD8+ T cells in SARS-CoV-2 infection. The cross-reactive responses intermediated by CD8+ T cells in unexposed cohorts are described. Additionally, the relevance of resident CD8+ T cells in the upper and lower airway during infection and CD8+ T-cell responses following vaccination are discussed, including recent worrisome breakthrough infections and variants of concerns (VOCs). Lastly, we explain the correlation between CD8+ T cells and COVID-19 severity. This review aids in a deeper comprehension of the association between CD8+ T cells and SARS-CoV-2 and broadens a vision for future exploration.
Collapse
Affiliation(s)
- Yuanting Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, USA
| | - Maria G. Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Fabio Cndotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kamel Benlagha
- Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Niels Olsen Saraiva Camara
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Junming Shi
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Lu Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Alexander Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
2
|
Sharma S, Roy D, Cherian S. In-silico evaluation of the T-cell based immune response against SARS-CoV-2 omicron variants. Sci Rep 2024; 14:25413. [PMID: 39455652 PMCID: PMC11511884 DOI: 10.1038/s41598-024-75658-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
During of COVID-19 pandemic, Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has continuously evolved, resulting in the emergence of several new variants of concerns (VOCs) with numerous mutations. These VOCs dominate in various regions due to increased transmissibility and antibody evasion, potentially reducing vaccine effectiveness. Nonetheless, it remains uncertain whether the recent SARS-CoV-2 VOCs have the ability to circumvent the T cell immunity elicited by either COVID-19 vaccination or natural infection. To address this, we conducted in-silico analysis to examine the impact of VOC-specific mutations at the epitope level and T cell cross-reactivity with the ancestral SARS-CoV-2. According to the in-silico investigation, T cell responses triggered by immunization or prior infections still recognize the variants in spite of mutations. These variants are expected to either maintain their dominant epitope HLA patterns or bind with new HLAs, unlike the epitopes of the ancestral strain. Our findings indicate that a significant proportion of immuno-dominant CD8 + and CD4 + epitopes are conserved across all the variants, implying that existing vaccines might maintain efficacy against new variations. However, further in-vitro and in-vivo studies are needed to validate the in-silico results and fully elucidate immune sensitivities to VOCs.
Collapse
Affiliation(s)
- Shivangi Sharma
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India
| | - Diya Roy
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India
| | - Sarah Cherian
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India.
| |
Collapse
|
3
|
Im KI, Kim N, Lee J, Oh UH, Lee HW, Lee DG, Min GJ, Lee R, Lee J, Kim S, Cho SG. SARS-CoV-2-Specific T-Cell as a Potent Therapeutic Strategy against Immune Evasion of Emerging COVID-19 Variants. Int J Mol Sci 2024; 25:10512. [PMID: 39408840 PMCID: PMC11477143 DOI: 10.3390/ijms251910512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing the protective abilities using virus-specific T cells (VSTs). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VSTs were generated by stimulating donor-derived peripheral blood mononuclear cells with spike, nucleocapsid, and membrane protein peptide mixtures. Phenotypic characterization, including T-cell receptor (TCR) vβ and pentamer analyses, was performed on the ex vivo-expanded cells. We infected human leukocyte antigen (HLA)-partially matched human Calu-3 cells with various authentic SARS-CoV-2 strains in a Biosafety Level 3 facility and co-cultured them with VSTs. VSTs exhibited a diverse TCR vβ repertoire, confirming their ability to target a broad range of SARS-CoV-2 antigens from both the ancestral and mutant strains, including Omicron BA.1 and BA.5. These ex vivo-expanded cells exhibited robust cytotoxicity and low alloreactivity against HLA-partially matched SARS-CoV-2-infected cells. Their cytotoxic effects were consistent across variants, targeting conserved spike and nucleocapsid epitopes. Our findings suggest that third-party partial HLA-matching VSTs could counter immune-escape mechanisms posed by emerging variants of concern.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Junseok Lee
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
| | - Ui-Hyeon Oh
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Hye-Won Lee
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Gi-June Min
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Raeseok Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jinah Lee
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
4
|
García-Pérez J, Borobia AM, Pérez-Olmeda M, Portolés A, Castaño L, Campins-Artí M, Bertrán MJ, Bermejo M, Arribas JR, López A, Ascaso-Del-Rio A, Arana-Arri E, Fuentes Camps I, Vilella A, Cascajero A, García-Morales MT, Castillo de la Osa M, Pérez Ingidua C, Lora D, Jiménez-Santana P, Pino-Rosa S, Gómez de la Cámara A, De La Torre-Tarazona E, Calonge E, Cruces R, Belda-Iniesta C, Alcamí J, Frías J, Carcas AJ, Díez-Fuertes F. Immunogenicity of a third dose with mRNA-vaccines in the ChAdOx1-S/BNT162b2 vaccination regimen against SARS-CoV-2 variants. iScience 2024; 27:110728. [PMID: 39286494 PMCID: PMC11404211 DOI: 10.1016/j.isci.2024.110728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/28/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
CombiVacS study has demonstrated a strong immune response of the heterologous ChAdOx1-S/BNT162b2 vaccine combination. The primary outcomes of the study were to assess the humoral immune response against SARS-CoV-2, 28 days after a third dose of a mRNA vaccine, in subjects that received a previous prime-boost scheme with ChAdOx1-S/BNT162b2. Secondary outcomes extended the study to 3 and 6 months. The third vaccine dose of mRNA-1273 in naive participants previously vaccinated with ChAdOx1-S/BNT162b2 regimen reached higher neutralizing antibodies titers against the variants of concern Delta and BA.1 lineage of Omicron compared with those receiving a third dose of BNT162b2 at day 28. These differences between BNT162b2 and mRNA-1273 arms were observed against the ancestral variant G614 at day 90. Suboptimal neutralizing response was observed against BQ.1.1, XBB.1.5/XBB.1.9, and JN.1 in a relevant proportion of individuals 180 days after the third dose, even after asymptomatic Omicron breakthrough infections. EudraCT (2021-001978-37); ClinicalTrials.gov (NCT04860739).
Collapse
Affiliation(s)
- Javier García-Pérez
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Alberto M Borobia
- Servicio de Farmacología Clínica, Hospital Universitario La Paz, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, IdiPAZ, 28046 Madrid, Spain
- Servicio de Medicina Interna, Departamento de Medicina, Facultad de Medicina, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, 28046 Madrid, Spain
| | - Mayte Pérez-Olmeda
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Antonio Portolés
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - Luis Castaño
- Cruces University Hospital, Bio-Bizkaia, UPV/EHU, CIBERDEM/CIBERER, Endo-ERN, Barakaldo, 48903 Vizcaya, Spain
| | - Magdalena Campins-Artí
- Servicio de Medicina Preventiva y Epidemiología, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - María Jesús Bertrán
- Servicio de Medicina Preventiva y Epidemiología, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Mercedes Bermejo
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - José Ramón Arribas
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Servicio de Medicina Interna, Departamento de Medicina, Facultad de Medicina, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, 28046 Madrid, Spain
| | - Andrea López
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Ana Ascaso-Del-Rio
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | - Eunate Arana-Arri
- Cruces University Hospital, Bio-Bizkaia, UPV/EHU, CIBERDEM/CIBERER, Endo-ERN, Barakaldo, 48903 Vizcaya, Spain
| | - Inmaculada Fuentes Camps
- Unidad de Soporte a la Investigación Clínica, Vall d'Hebron Institut de Recerca, Servicio de Farmacología Clínica, Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Anna Vilella
- Servicio de Medicina Preventiva y Epidemiología, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Almudena Cascajero
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | | | - María Castillo de la Osa
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Carla Pérez Ingidua
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain
| | - David Lora
- Spanish Clinical Research Network - SCReN - ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario 12 de octubre, 28041 Madrid, Spain
- Facultad de Estudios Estadísticos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Paloma Jiménez-Santana
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Silvia Pino-Rosa
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Agustín Gómez de la Cámara
- Instituto de Investigación Sanitaria del Hospital Universitario 12 de octubre, 28041 Madrid, Spain
- Facultad de Estudios Estadísticos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Erick De La Torre-Tarazona
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Esther Calonge
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Raquel Cruces
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | | | - José Alcamí
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| | - Jesús Frías
- Servicio de Farmacología Clínica, Hospital Universitario La Paz, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, IdiPAZ, 28046 Madrid, Spain
| | - Antonio J Carcas
- Servicio de Farmacología Clínica, Hospital Universitario La Paz, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, IdiPAZ, 28046 Madrid, Spain
| | - Francisco Díez-Fuertes
- Unidad de Inmunopatología del SIDA, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28222 Madrid, Spain
| |
Collapse
|
5
|
Tsagkli P, Geropeppa M, Papadatou I, Spoulou V. Hybrid Immunity against SARS-CoV-2 Variants: A Narrative Review of the Literature. Vaccines (Basel) 2024; 12:1051. [PMID: 39340081 PMCID: PMC11436074 DOI: 10.3390/vaccines12091051] [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: 08/11/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
The emergence of SARS-CoV-2 led to a global health crisis and the burden of the disease continues to persist. The rapid development and emergency authorization of various vaccines, including mRNA-based vaccines, played a pivotal role in mitigating severe illness and mortality. However, rapid viral mutations, leading to several variants of concern, challenged vaccine effectiveness, particularly concerning immune evasion. Research on immunity, both from natural infection and vaccination, revealed that while neutralizing antibodies provide protection against infection, their effect is short-lived. The primary defense against severe COVID-19 is derived from the cellular immune response. Hybrid immunity, developed from a combination of natural infection and vaccination, offers enhanced protection, with convalescent vaccinated individuals showing significantly higher levels of neutralizing antibodies. As SARS-CoV-2 continues to evolve, understanding the durability and breadth of hybrid immunity becomes crucial. This narrative review examines the latest data on humoral and cellular immunity from both natural infection and vaccination, discussing how hybrid immunity could inform and optimize future vaccination strategies in the ongoing battle against COVID-19 and in fear of a new pandemic.
Collapse
Affiliation(s)
- Panagiota Tsagkli
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| | - Maria Geropeppa
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| | - Ioanna Papadatou
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory and Infectious Diseases Department "MAKKA", First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Athens Medical School, 11527 Athens, Greece
| |
Collapse
|
6
|
Yao D, Patel RS, Lam A, Glover Q, Srinivasan C, Herchen A, Ritchie B, Agrawal B. Antibody Responses in SARS-CoV-2-Exposed and/or Vaccinated Individuals Target Conserved Epitopes from Multiple CoV-2 Antigens. Int J Mol Sci 2024; 25:9814. [PMID: 39337303 PMCID: PMC11432605 DOI: 10.3390/ijms25189814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
There is a need to investigate novel strategies in order to create an effective, broadly protective vaccine for current and future severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreaks. The currently available vaccines demonstrate compromised efficacy against emerging SARS-CoV-2 variants of concern (VOCs), short-lived immunity, and susceptibility to immune imprinting due to frequent boosting practices. In this study, we examined the specificity of cross-reactive IgG antibody responses in mRNA-vaccinated, AstraZeneca-vaccinated, and unvaccinated donors to identify potentially conserved, cross-reactive epitopes to target in order to create a broadly protective SARS-CoV-2 vaccine. Our study provides evidence for cross-reactive IgG antibodies specific to eight different spike (S) variants. Furthermore, the specificities of these cross-variant IgG antibody titers were associated to some extent with spike S1- and S2-subunit-derived epitopes P1 and P2, respectively. In addition, nucleocapsid (N)- and membrane (M)-specific IgG antibody titers correlated with N- and M-derived epitopes conserved across beta-CoVs, P3-7. This study reveals conserved epitopes of viral antigens, targeted by natural and/or vaccine-induced human immunity, for future designs of next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- David Yao
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| | - Raj S. Patel
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| | - Adrien Lam
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| | - Quarshie Glover
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Cindy Srinivasan
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Alex Herchen
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Bruce Ritchie
- Department of Medicine, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (Q.G.); (C.S.); (A.H.); (B.R.)
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada; (D.Y.); (R.S.P.); (A.L.)
| |
Collapse
|
7
|
McMahon WC, Kwatra G, Izu A, Jones SA, Mbele NJ, Jafta N, Lala R, Shalekoff S, Tiemessen CT, Madhi SA, Nunes MC. T-cell responses to ancestral SARS-CoV-2 and Omicron variant among unvaccinated pregnant and postpartum women living with and without HIV in South Africa. Sci Rep 2024; 14:20348. [PMID: 39223211 PMCID: PMC11369237 DOI: 10.1038/s41598-024-70725-8] [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: 12/18/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
SARS-CoV-2 cell-mediated immunity remains understudied during pregnancy in unvaccinated Black African women living with HIV (WLWH) from low- and middle-income countries. We investigated SARS-CoV-2-specific T-cell responses 1 month following infection in 24 HIV-uninfected women and 15 WLWH at any stage during pregnancy or postpartum. The full-length spike (FLS) glycoprotein and nucleocapsid (N) protein of wild-type (WT) SARS-CoV-2, as well as mutated spike protein regions found in the Omicron variant (B.1.1.529) were targeted by flow cytometry. WT-specific CD4+ and CD8+ T cells elicited similar FLS- and N-specific responses in HIV-uninfected women and WLWH. SARS-CoV-2-specific T-lymphocytes were predominantly TNF-α monofunctional in pregnant and postpartum women living with and without HIV, with fever cells producing either IFN-γ or IL-2. Furthermore, T-cell responses were unaffected by Omicron-specific spike mutations as similar responses between Omicron and the ancestral virus were detected for CD4+ and CD8+ T cells. Our results collectively demonstrate comparable T-cell responses between WLWH on antiretroviral therapy and HIV-uninfected pregnant and postpartum women who were naïve to Covid-19 vaccination. Additionally, we show that T cells from women infected with the ancestral virus, Beta variant (B.1.351), or Delta variant (B.1.617.2) can cross-recognize Omicron, suggesting an overall preservation of T-cell immunity.
Collapse
Affiliation(s)
- William C McMahon
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Research Chair Initiative in Vaccine Preventable Diseases, Department of Science and Innovation/National Research Foundation, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA.
- Department of Clinical Microbiology, Christian Medical College, Vellore, India.
| | - Alane Izu
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Stephanie A Jones
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nkululeko J Mbele
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nwabisa Jafta
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rushil Lala
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sharon Shalekoff
- A Division of the National Health Laboratory Service, Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline T Tiemessen
- A Division of the National Health Laboratory Service, Centre for HIV and STIs, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marta C Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Research Chair Initiative in Vaccine Preventable Diseases, Department of Science and Innovation/National Research Foundation, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Center of Excellence in Respiratory Pathogens, Hospices Civils de Lyon, and Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
8
|
Serdyuk YV, Zornikova KV, Dianov DV, Ivanova NO, Davydova VD, Fefelova EI, Nenasheva TA, Sheetikov SA, Bogolyubova AV. T-Cell Receptors Cross-Reactive to Coronaviral Epitopes Homologous to the SPR Peptide. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1631-1642. [PMID: 39418521 DOI: 10.1134/s0006297924090098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024]
Abstract
The COVID-19 pandemic caused by the rapid spread of the novel coronavirus SARS-CoV-2, has promoted an interest in studying the T-cell immune response. It was found that the polyclonal and cross-reactive T-cell response against seasonal coronaviruses and other SARS-CoV-2 strains reduced disease severity. We investigated the immunodominant T-cell epitope SPRWYFYYYL from the nucleocapsid protein of SARS-CoV-2. The immune response to this epitope is characterized by the formation of highly homologous (convergent) receptors that have been found in the T-cell receptor (TCR) repertoires of different individuals. This epitope belongs to a group of highly conserved peptides that are rarely mutated in novel SARS-CoV-2 strains and are homologous to the epitopes of seasonal coronaviruses. It has been suggested that the cross-reactive response to homologous peptides contributes to the reduction of COVID-19 severity. However, some investigators have questioned this hypothesis, suggesting that the low affinity of the cross-reactive receptors reduces the strength of the immune response. The aim of this study was to evaluate the effect of amino acid substitutions in the SPR epitope on its binding affinity to specific TCRs. For this, we performed antigen-dependent cellular expansions were performed using samples from four COVID-19-transfected donors and sequenced their TCR repertoires. The resulting SPR-specific repertoire of β-chains in TCRs had a greater sequence diversity than the repertoire of α-chains. However, the TCR repertoires of all four donors contained public receptors, three of which were cloned and used to generate the Jurkat E6-1 TPR cell line. Only one of these receptors was activated by the SPR peptide and recognized with the same affinity by its mutant homologue LPRWYFYYY from seasonal coronaviruses. This indicates that the presence of the mutation did not affect the strength of the immune response, which may explain why the cross-reactive response to the SPR epitope is so frequent and contributes positively to COVID-19 infection.
Collapse
Affiliation(s)
- Yana V Serdyuk
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ksenia V Zornikova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Dmitry V Dianov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Nataliia O Ivanova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Vassa D Davydova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ekaterina I Fefelova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Tatiana A Nenasheva
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Saveliy A Sheetikov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Apollinariya V Bogolyubova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| |
Collapse
|
9
|
Liu X, Ng WH, Zusinaite E, Freitas J, Taylor A, Yerragunta V, Aavula SM, Gorriparthi S, Ponsekaran S, Bonda RL, Mani P, Nimmagadda SV, Wang S, Lello LS, Zaid A, Dua U, Taft-Benz SA, Anderson E, Baxter VK, Sarkar S, Ling ZL, Ashhurst TM, Cheng SMS, Pattnaik P, Kanakasapapathy AK, Baric RS, Burt FJ, Peiris M, Heise MT, King NJC, Merits A, Lingala R, Mahalingam S. A single-dose intranasal live-attenuated codon deoptimized vaccine provides broad protection against SARS-CoV-2 and its variants. Nat Commun 2024; 15:7225. [PMID: 39187479 PMCID: PMC11347628 DOI: 10.1038/s41467-024-51535-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19) continues its significant health and economic impact globally. Despite the success of spike-protein vaccines in preventing severe disease, long-lasting protection against emerging variants and the prevention of breakthrough infections and transmission remain elusive. We generate an intranasal live-attenuated SARS-CoV-2 vaccine, CDO-7N-1, using codon deoptimization. CDO-7N-1 shows highly attenuated replication and minimal or no lung pathology in vivo over multiple passages. It induces robust mucosal and systemic neutralizing antibody and T-cell subset responses, in mice (female K18-hACE2 and male HFH4-hACE2 mice), hamsters, and macaques triggered by a single immunization. Mice and hamsters vaccinated with CDO-7N-1 are protected from challenge with wild-type (WT) SARS-CoV-2 and other variants of concern. Serum from vaccinated animals neutralizes WT SARS-CoV-2, variants of concern (beta and delta), variants of interest (omicron XBB.1.5) and SARS-CoV-1. Antibody responses are sustained and enhanced by repeated immunization or infection with WT SARS-CoV-2. Immunity against all SARS-CoV-2 proteins by CDO-7N-1 should improve efficacy against future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Xiang Liu
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Wern Hann Ng
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Eva Zusinaite
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia
| | - Joseph Freitas
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Adam Taylor
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Venugopal Yerragunta
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Shukra Madhaha Aavula
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Sambaiah Gorriparthi
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Santhakumar Ponsekaran
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Rama Lakshmi Bonda
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Priyanka Mani
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Sridevi V Nimmagadda
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Sainan Wang
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia
| | | | - Ali Zaid
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Ujjwal Dua
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- Griffith Enterprise, Griffith University, Brisbane, QLD, Australia
| | - Sharon A Taft-Benz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth Anderson
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria K Baxter
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sanjay Sarkar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zheng L Ling
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Thomas M Ashhurst
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Cytometry Core Research Facility, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
| | - Samuel M S Cheng
- School of Public Health, The University of Hong Kong, Special Administrative Region of Hong Kong, Hong Kong, China
| | - Priyabrata Pattnaik
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | | | - Ralph S Baric
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Felicity J Burt
- Division of Virology, National Health Laboratory Service and Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Malik Peiris
- School of Public Health, The University of Hong Kong, Special Administrative Region of Hong Kong, Hong Kong, China
- HKU-Pasteur Research Pole, The University of Hong Kong, Special Administrative Region of Hong Kong, Hong Kong, China
| | - Mark T Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholas J C King
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Viral Immunopathology Laboratory, The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Nano, The University of Sydney, Sydney, NSW 2006, Australia
| | - Andres Merits
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia
| | - Rajendra Lingala
- Indian Immunologicals Ltd (IIL), Rakshapuram, Gachibowli Post, Hyderabad, 500032, Telangana, India
| | - Suresh Mahalingam
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia.
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
10
|
Leno-Duran E, Serrano-Conde E, Salas-Rodríguez A, Salcedo-Bellido I, Barrios-Rodríguez R, Fuentes A, Viñuela L, García F, Requena P. Evaluation of inflammatory biomarkers and their association with anti-SARS-CoV-2 antibody titers in healthcare workers vaccinated with BNT162B2. Front Immunol 2024; 15:1447317. [PMID: 39247198 PMCID: PMC11377239 DOI: 10.3389/fimmu.2024.1447317] [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: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Vaccine-induced immunity against COVID-19 generates antibody and lymphocyte responses. However, variability in antibody titers has been observed after vaccination, and the determinants of a better response should be studied. The main objective of this investigation was to analyze the inflammatory biomarker response induced in healthcare workers vaccinated with BNT162b2, and its association with anti-Spike (a SARS-CoV-2 antigen) antibodies measured throughout a 1-year follow-up. Methods Anti-spike antibodies and 92 biomarkers were analyzed in serum, along with socio-demographic and clinical variables collected by interview or exploration. Results In our study, four biomarkers (ADA, IL-17C, CCL25 and CD8α) increased their expression after the first vaccine dose; and 8 others (uPA, IL-18R1, EN-RAGE, CASP-8, MCP-2, TNFβ, CD5 and CXCL10) decreased their expression. Age, body mass index (BMI), smoking, alcohol consumption, and prevalent diseases were associated with some of these biomarkers. Furthermore, higher baseline levels of T-cell surface glycoprotein CD6 and hepatocyte growth factor (HGF) were associated with lower mean antibody titers at follow-up, while levels of monocyte chemotactic protein 2 (MCP-2) had a positive association with antibody levels. Age and BMI were positively related to baseline levels of MCP-2 (β=0.02, 95%CI 0.00-0.04, p=0.036) and HGF (β=0.03, 95%CI 0.00-0.06, p=0.039), respectively. Conclusion Our findings indicate that primary BNT162b2 vaccination had a positive effect on the levels of several biomarkers related to T cell function, and a negative one on some others related to cancer or inflammatory processes. In addition, a higher level of MCP-2 and lower levels of HGF and CD6 were found to be associated with higher anti-Spike antibody titer following vaccination.
Collapse
Affiliation(s)
- Ester Leno-Duran
- Universidad de Granada, Departamento de Obstetricia y Ginecología, Granada, Spain
| | - Esther Serrano-Conde
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Ana Salas-Rodríguez
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
| | - Inmaculada Salcedo-Bellido
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Rocío Barrios-Rodríguez
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Ana Fuentes
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Laura Viñuela
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Federico García
- Servicio de Microbiología, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Pilar Requena
- Universidad de Granada, Departamento de Medicina Preventiva y Salud Pública, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
11
|
De Biasi S, Ciobanu AL, Santacroce E, Lo Tartaro D, Degliesposti G, D’Angerio M, Leccese M, Cardi M, Trenti T, Cuccorese M, Gibellini L, Ferraro D, Cossarizza A. SARS-CoV-2 Vaccination Responses in Anti-CD20-Treated Progressive Multiple Sclerosis Patients Show Immunosenescence in Antigen-Specific B and T Cells. Vaccines (Basel) 2024; 12:924. [PMID: 39204047 PMCID: PMC11360119 DOI: 10.3390/vaccines12080924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) shows that inflammation starts early and progresses with age. B cells play a central role in this process, contributing to cytokine production, defective regulatory functions, and abnormal immunoglobulin production, even in the central nervous system. Anti-CD20 (aCD20) therapies, which deplete CD20+ B cells, are largely used in the treatment of both relapsing remitting (RR) and progressive (PR) forms of MS. Although effective against MS symptoms and lesions detectable by magnetic resonance imaging, aCD20 therapies can reduce the immune response to COVID-19 vaccination. By using high-parameter flow cytometry, we examined the antigen-specific (Ag+) immune response six months post-third COVID-19 mRNA vaccination in MS patients with RR and PR forms on aCD20 therapy. Despite lower Ag+ B cell responses and lower levels of anti-SARS-CoV2, both total and neutralizing antibodies, RR and PR patients developed strong Ag+ T cell responses. We observed similar percentages and numbers of Ag+ CD4+ T cells and a high proportion of Ag+ CD8+ T cells, with slight differences in T cell phenotype and functionality; this, however, suggested the presence of differences in immune responses driven by age and disease severity.
Collapse
Affiliation(s)
- Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Gianluca Degliesposti
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Miriam D’Angerio
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Maristella Leccese
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Martina Cardi
- AOU Policlinico di Modena, Neurology Unit, Department of Biomedical, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Tommaso Trenti
- AOU Policlinico di Modena, Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, 41124 Modena, Italy
| | - Michela Cuccorese
- AOU Policlinico di Modena, Diagnostic Hematology and Clinical Genomics, Department of Laboratory Medicine and Pathology, 41124 Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
| | - Diana Ferraro
- AOU Policlinico di Modena, Neurology Unit, Department of Biomedical, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy (A.C.)
- National Institute for Cardiovascular Research, 40126 Bologna, Italy
| |
Collapse
|
12
|
Kister I, Curtin R, Piquet AL, Borko T, Pei J, Banbury BL, Bacon TE, Kim A, Tuen M, Velmurugu Y, Nyovanie S, Selva S, Samanovic MI, Mulligan MJ, Patskovsky Y, Priest J, Cabatingan M, Winger RC, Krogsgaard M, Silverman GJ. Longitudinal study of immunity to SARS-CoV2 in ocrelizumab-treated MS patients up to 2 years after COVID-19 vaccination. Ann Clin Transl Neurol 2024; 11:1750-1764. [PMID: 38713096 PMCID: PMC11251481 DOI: 10.1002/acn3.52081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 05/08/2024] Open
Abstract
OBJECTIVES (1) To plot the trajectory of humoral and cellular immune responses to the primary (two-dose) COVID-19 mRNA series and the third/booster dose in B-cell-depleted multiple sclerosis (MS) patients up to 2 years post-vaccination; (2) to identify predictors of immune responses to vaccination; and (3) to assess the impact of intercurrent COVID-19 infections on SARS CoV-2-specific immunity. METHODS Sixty ocrelizumab-treated MS patients were enrolled from NYU (New York) and University of Colorado (Anschutz) MS Centers. Samples were collected pre-vaccination, and then 4, 12, 24, and 48 weeks post-primary series, and 4, 12, 24, and 48 weeks post-booster. Binding anti-Spike antibody responses were assessed with multiplex bead-based immunoassay (MBI) and electrochemiluminescence (Elecsys®, Roche Diagnostics), and neutralizing antibody responses with live-virus immunofluorescence-based microneutralization assay. Spike-specific cellular responses were assessed with IFNγ/IL-2 ELISpot (Invitrogen) and, in a subset, by sequencing complementarity determining regions (CDR)-3 within T-cell receptors (Adaptive Biotechnologies). A linear mixed-effect model was used to compare antibody and cytokine levels across time points. Multivariate analyses identified predictors of immune responses. RESULTS The primary vaccination induced an 11- to 208-fold increase in binding and neutralizing antibody levels and a 3- to 4-fold increase in IFNγ/IL-2 responses, followed by a modest decline in antibody but not cytokine responses. Booster dose induced a further 3- to 5-fold increase in binding antibodies and 4- to 5-fold increase in IFNγ/IL-2, which were maintained for up to 1 year. Infections had a variable impact on immunity. INTERPRETATION Humoral and cellular benefits of COVID-19 vaccination in B-cell-depleted MS patients were sustained for up to 2 years when booster doses were administered.
Collapse
MESH Headings
- Humans
- COVID-19/immunology
- COVID-19/prevention & control
- Male
- Female
- Middle Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/administration & dosage
- Adult
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Longitudinal Studies
- SARS-CoV-2/immunology
- Multiple Sclerosis/immunology
- Multiple Sclerosis/drug therapy
- Antibodies, Viral/blood
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Immunity, Cellular/drug effects
- Vaccination
- Immunity, Humoral/drug effects
- Immunity, Humoral/immunology
- BNT162 Vaccine/administration & dosage
- BNT162 Vaccine/immunology
Collapse
Affiliation(s)
- Ilya Kister
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Ryan Curtin
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Amanda L. Piquet
- Rocky Mountain MS CenterUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Tyler Borko
- Rocky Mountain MS CenterUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Jinglan Pei
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | | | - Tamar E. Bacon
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Angie Kim
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Michael Tuen
- NYU Langone Vaccine Center and Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Yogambigai Velmurugu
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Samantha Nyovanie
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Sean Selva
- Rocky Mountain MS CenterUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Marie I. Samanovic
- NYU Langone Vaccine Center and Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Mark J. Mulligan
- NYU Langone Vaccine Center and Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Yury Patskovsky
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | | | | | | | - Michelle Krogsgaard
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Gregg J. Silverman
- Division of Rheumatology, Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| |
Collapse
|
13
|
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] [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
|
14
|
Richards KA, Changrob S, Thomas PG, Wilson PC, Sant AJ. Lack of memory recall in human CD4 T cells elicited by the first encounter with SARS-CoV-2. iScience 2024; 27:109992. [PMID: 38868209 PMCID: PMC11166706 DOI: 10.1016/j.isci.2024.109992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/11/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
The studies reported here focus on the impact of pre-existing CD4 T cell immunity on the first encounter with SARS-CoV-2. They leverage PBMC samples from plasma donors collected after a first SARS-CoV-2 infection, prior to vaccine availability and compared to samples collected prior to the emergence of SARS-CoV-2. Analysis of CD4 T cell specificity across the entire SARS-CoV-2 proteome revealed that the recognition of SARS-CoV-2-derived epitopes by CD4 memory cells prior to the pandemic are enriched for reactivity toward non-structural proteins conserved across endemic CoV strains. However, CD4 T cells after primary infection with SARS-CoV-2 focus on epitopes from structural proteins. We observed little evidence for preferential recall to epitopes conserved between SARS-CoV-2 and seasonal CoV, a finding confirmed through use of selectively curated conserved and SARS-unique peptides. Our data suggest that SARS-CoV-2 CD4 T cells elicited by the first infection are primarily established from the naive CD4 T cell pool.
Collapse
Affiliation(s)
- Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Siriruk Changrob
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Patrick C. Wilson
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
15
|
Cheng YL, Chang SS, Chao CH, Chen PT, Lin YL, Syu GD, Lee NY, Chen PL, Ko WC, Ho TS. Optimizing SARS-CoV-2 vaccine responses in kidney transplant recipients: an urgent need. Microbiol Spectr 2024; 12:e0000424. [PMID: 38747636 PMCID: PMC11237705 DOI: 10.1128/spectrum.00004-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024] Open
Abstract
Kidney transplant recipients (KTRs) have been identified as a population at increased risk for severe SARS-CoV-2 infection outcomes. This study focused on understanding the immune response of KTRs post-vaccination, specifically examining both serological and cellular responses to the SARS-CoV-2 vaccine. Thirteen individuals, including seven KTRs and six healthy donors, were evaluated for antibody levels and T cell responses post-vaccination. The study revealed that KTRs had significantly lower serological responses, including reduced anti-receptor binding domain (RBD) binding antibodies and neutralizing antibodies against the Wuhan, Delta, and Omicron BA.2 strains. Additionally, KTRs demonstrated weaker CD8 T cell cytotoxic responses and lower Th1 cytokine secretion, particularly IFN-γ, after stimulation with variant spike peptide pools. These findings highlight the compromised immunity in KTRs post-vaccination and underscore the need for tailored strategies to bolster immune responses in this vulnerable group. Further investigations are warranted into the mechanisms underlying reduced vaccine efficacy in KTRs and potential therapeutic interventions. IMPORTANCE Some studies have revealed that KTRs had lower serological response against SARS-CoV-2 than healthy people. Nevertheless, limited studies investigate the cellular response against SARS-CoV-2 in KTRs receiving SARS-CoV-2 vaccines. Here, we found that KTRs have lower serological and cellular responses. Moreover, we found that KTRs had a significantly lower IFN-γ secretion than healthy individuals when their PBMCs were stimulated with SARS-CoV-2 spike peptide pools. Thus, our findings suggested that additional strategies are needed to enhance KTR immunity triggered by the vaccine.
Collapse
Affiliation(s)
- Yi-Ling Cheng
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shen-Shin Chang
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiao-Hsuan Chao
- Department of Medical Laboratory and Regenerative Medicine, MacKay Medical College, New Taipei, Taiwan
| | - Po-Ta Chen
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Lan Lin
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Guan-Da Syu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Nan-Yao Lee
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Po-Lin Chen
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzong-Shiann Ho
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Pediatrics, National Cheng Kung University Hospital Dou-Liou Branch, College of Medicine, National Cheng Kung University, Yunlin, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
16
|
Kim GJ, Elnaggar JH, Varnado M, Feehan AK, Tauzier D, Rose R, Lamers SL, Sevalia M, Nicholas N, Gravois E, Fort D, Crabtree JS, Miele L. A bioinformatic analysis of T-cell epitope diversity in SARS-CoV-2 variants: association with COVID-19 clinical severity in the United States population. Front Immunol 2024; 15:1357731. [PMID: 38784379 PMCID: PMC11112498 DOI: 10.3389/fimmu.2024.1357731] [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: 12/18/2023] [Accepted: 04/05/2024] [Indexed: 05/25/2024] Open
Abstract
Long-term immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires the identification of T-cell epitopes affecting host immunogenicity. In this computational study, we explored the CD8+ epitope diversity estimated in 27 of the most common HLA-A and HLA-B alleles, representing most of the United States population. Analysis of 16 SARS-CoV-2 variants [B.1, Alpha (B.1.1.7), five Delta (AY.100, AY.25, AY.3, AY.3.1, AY.44), and nine Omicron (BA.1, BA.1.1, BA.2, BA.4, BA.5, BQ.1, BQ.1.1, XBB.1, XBB.1.5)] in analyzed MHC class I alleles revealed that SARS-CoV-2 CD8+ epitope conservation was estimated at 87.6%-96.5% in spike (S), 92.5%-99.6% in membrane (M), and 94.6%-99% in nucleocapsid (N). As the virus mutated, an increasing proportion of S epitopes experienced reduced predicted binding affinity: 70% of Omicron BQ.1-XBB.1.5 S epitopes experienced decreased predicted binding, as compared with ~3% and ~15% in the earlier strains Delta AY.100-AY.44 and Omicron BA.1-BA.5, respectively. Additionally, we identified several novel candidate HLA alleles that may be more susceptible to severe disease, notably HLA-A*32:01, HLA-A*26:01, and HLA-B*53:01, and relatively protected from disease, such as HLA-A*31:01, HLA-B*40:01, HLA-B*44:03, and HLA-B*57:01. Our findings support the hypothesis that viral genetic variation affecting CD8 T-cell epitope immunogenicity contributes to determining the clinical severity of acute COVID-19. Achieving long-term COVID-19 immunity will require an understanding of the relationship between T cells, SARS-CoV-2 variants, and host MHC class I genetics. This project is one of the first to explore the SARS-CoV-2 CD8+ epitope diversity that putatively impacts much of the United States population.
Collapse
Affiliation(s)
- Grace J. Kim
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jacob H. Elnaggar
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Department of Microbiology, Immunology, and Parasitology, Lousiana State University Health Sciences Center (LSUHSC), New Orleans, LA, United States
| | - Mallory Varnado
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Amy K. Feehan
- Research and Development, Oschner Medical Center, New Orleans, LA, United States
| | - Darlene Tauzier
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Rebecca Rose
- Research and Development, BioInfoExperts, LLC, Thibodaux, LA, United States
| | - Susanna L. Lamers
- Research and Development, BioInfoExperts, LLC, Thibodaux, LA, United States
| | - Maya Sevalia
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Najah Nicholas
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Elizabeth Gravois
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Daniel Fort
- Research and Development, Oschner Medical Center, New Orleans, LA, United States
| | - Judy S. Crabtree
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
17
|
Brummelman J, Suárez-Hernández S, de Rond L, Bogaard-van Maurik M, Molenaar P, van Wijlen E, Oomen D, Beckers L, Rots NY, van Beek J, Nicolaie MA, van Els CACM, Boer MC, Kaaijk P, Buisman AM, de Wit J. Distinct T cell responsiveness to different COVID-19 vaccines and cross-reactivity to SARS-CoV-2 variants with age and CMV status. Front Immunol 2024; 15:1392477. [PMID: 38774878 PMCID: PMC11106399 DOI: 10.3389/fimmu.2024.1392477] [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: 02/27/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Accumulating evidence indicates the importance of T cell immunity in vaccination-induced protection against severe COVID-19 disease, especially against SARS-CoV-2 Variants-of-Concern (VOCs) that more readily escape from recognition by neutralizing antibodies. However, there is limited knowledge on the T cell responses across different age groups and the impact of CMV status after primary and booster vaccination with different vaccine combinations. Moreover, it remains unclear whether age has an effect on the ability of T cells to cross-react against VOCs. Methods Therefore, we interrogated the Spike-specific T cell responses in healthy adults of the Dutch population across different ages, whom received different vaccine types for the primary series and/or booster vaccination, using IFNɣ ELISpot. Cells were stimulated with overlapping peptide pools of the ancestral Spike protein and different VOCs. Results Robust Spike-specific T cell responses were detected in the vast majority of participants upon the primary vaccination series, regardless of the vaccine type (i.e. BNT162b2, mRNA-1273, ChAdOx1 nCoV-19, or Ad26.COV2.S). Clearly, in the 70+ age group, responses were overall lower and showed more variation compared to younger age groups. Only in CMV-seropositive older adults (>70y) there was a significant inverse relation of age with T cell responses. Although T cell responses increased in all age groups after booster vaccination, Spike-specific T cell frequencies remained lower in the 70+ age group. Regardless of age or CMV status, primary mRNA-1273 vaccination followed by BNT162b2 booster vaccination showed limited booster effect compared to the BNT162b2/BNT162b2 or BNT162b2/mRNA-1273 primary-booster regimen. A modest reduction in cross-reactivity to the Alpha, Delta and Omicron BA.1, but not the Beta or Gamma variant, was observed after primary vaccination. Discussion Together, this study shows that age, CMV status, but also the primary-booster vaccination regimen influence the height of the vaccination-induced Spike-specific T cell response, but did not impact the VOC cross-reactivity.
Collapse
Affiliation(s)
- Jolanda Brummelman
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Sara Suárez-Hernández
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lia de Rond
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Marjan Bogaard-van Maurik
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Petra Molenaar
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Emma van Wijlen
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Debbie Oomen
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lisa Beckers
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Nynke Y. Rots
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Mioara A. Nicolaie
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Cécile A. C. M. van Els
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Mardi C. Boer
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Patricia Kaaijk
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Anne-Marie Buisman
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Jelle de Wit
- Center for Infectious Disease Control, Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
18
|
Hou Q, Jiang J, Na K, Zhang X, Liu D, Jing Q, Yan C, Han Y. Potential therapeutic targets for COVID-19 complicated with pulmonary hypertension: a bioinformatics and early validation study. Sci Rep 2024; 14:9294. [PMID: 38653779 DOI: 10.1038/s41598-024-60113-7] [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: 12/12/2023] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
Coronavirus disease (COVID-19) and pulmonary hypertension (PH) are closely correlated. However, the mechanism is still poorly understood. In this article, we analyzed the molecular action network driving the emergence of this event. Two datasets (GSE113439 and GSE147507) from the GEO database were used for the identification of differentially expressed genes (DEGs).Common DEGs were selected by VennDiagram and their enrichment in biological pathways was analyzed. Candidate gene biomarkers were selected using three different machine-learning algorithms (SVM-RFE, LASSO, RF).The diagnostic efficacy of these foundational genes was validated using independent datasets. Eventually, we validated molecular docking and medication prediction. We found 62 common DEGs, including several ones that could be enriched for Immune Response and Inflammation. Two DEGs (SELE and CCL20) could be identified by machine-learning algorithms. They performed well in diagnostic tests on independent datasets. In particular, we observed an upregulation of functions associated with the adaptive immune response, the leukocyte-lymphocyte-driven immunological response, and the proinflammatory response. Moreover, by ssGSEA, natural killer T cells, activated dendritic cells, activated CD4 T cells, neutrophils, and plasmacytoid dendritic cells were correlated with COVID-19 and PH, with SELE and CCL20 showing the strongest correlation with dendritic cells. Potential therapeutic compounds like FENRETI-NIDE, AFLATOXIN B1 and 1-nitropyrene were predicted. Further molecular docking and molecular dynamics simulations showed that 1-nitropyrene had the most stable binding with SELE and CCL20.The findings indicated that SELE and CCL20 were identified as novel diagnostic biomarkers for COVID-19 complicated with PH, and the target of these two key genes, FENRETI-NIDE and 1-nitropyrene, was predicted to be a potential therapeutic target, thus providing new insights into the prediction and treatment of COVID-19 complicated with PH in clinical practice.
Collapse
Affiliation(s)
- Qingbin Hou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jinping Jiang
- Department of Cardiology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Kun Na
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaolin Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Quanmin Jing
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
19
|
Montmaneix-Engels F, Dimeglio C, Staes L, Da Silva I, Porcheron M, Jougla I, Hérin F, Izopet J. Study of the cellular and humoral immune responses to SARS-CoV-2 vaccination. Heliyon 2024; 10:e29116. [PMID: 38601689 PMCID: PMC11004869 DOI: 10.1016/j.heliyon.2024.e29116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024] Open
Abstract
Our understanding of cellular immunity in response to COVID-19 infection or vaccination is limited because of less commonly used techniques. We investigated both the cellular and humoral immune responses before and after the administration of a third dose of the SARS-CoV-2 vaccine among a group of healthcare workers. Cellular immunity was evaluated using the VIDAS interferon-gamma (IFNγ) RUO test, which enables automated measurement of IFNγ levels after stimulating peripheral blood lymphocytes. Booster doses significantly enhanced both cellular and humoral immunity. Concerning cellular response, the booster dose increased the percentage of positive IFNγ release assay (IGRA) results but no difference in IFNγ release was found. The cellular response was not associated with protection against SARS-CoV-2 infection. Interestingly, vaccinated and infected healthcare workers exhibited the highest levels of anti-spike and neutralizing antibodies. In conclusion, the IGRA is a simple method for measuring cellular immune responses after vaccination. However, its usefulness as a complement to the study of humoral responses is yet to be demonstrated in future research.
Collapse
Affiliation(s)
- Faustine Montmaneix-Engels
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
| | - Chloé Dimeglio
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Laeticia Staes
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Da Silva
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Marion Porcheron
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| | - Isabelle Jougla
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
| | - Fabrice Hérin
- Occupational Diseases Department, Toulouse University Hospital, 31000, Toulouse, France
- UMR1295, Joint Research Unit INSERM- University Toulouse III Paul Sabatier, Centre for Epidemiology and Research in Population Health Unit (CERPOP), 31000, Toulouse, France
| | - Jacques Izopet
- INSERM UMR1291-CNRS UMR5051-University Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases, 31300, Toulouse, France
- Toulouse III Paul Sabatier University, 31062, Toulouse, France
- CHU Toulouse, Purpan Hospital, Virology Laboratory, 31300, Toulouse, France
| |
Collapse
|
20
|
Kristensen NP, Dionisio E, Bentzen AK, Tamhane T, Kemming JS, Nos G, Voss LF, Hansen UK, Lauer GM, Hadrup SR. Simultaneous analysis of pMHC binding and reactivity unveils virus-specific CD8 T cell immunity to a concise epitope set. SCIENCE ADVANCES 2024; 10:eadm8951. [PMID: 38608022 PMCID: PMC11014448 DOI: 10.1126/sciadv.adm8951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
CD8 T cells provide immunity to virus infection through recognition of epitopes presented by peptide major histocompatibility complexes (pMHCs). To establish a concise panel of widely recognized T cell epitopes from common viruses, we combined analysis of TCR down-regulation upon stimulation with epitope-specific enumeration based on barcode-labeled pMHC multimers. We assess CD8 T cell binding and reactivity for 929 previously reported epitopes in the context of 1 of 25 HLA alleles representing 29 viruses. The prevalence and magnitude of CD8 T cell responses were evaluated in 48 donors and reported along with 137 frequently recognized virus epitopes, many of which were underrepresented in the public domain. Eighty-four percent of epitope-specific CD8 T cell populations demonstrated reactivity to peptide stimulation, which was associated with effector and long-term memory phenotypes. Conversely, nonreactive T cell populations were associated primarily with naive phenotypes. Our analysis provides a reference map of epitopes for characterizing CD8 T cell responses toward common human virus infections.
Collapse
Affiliation(s)
- Nikolaj Pagh Kristensen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Edoardo Dionisio
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Amalie Kai Bentzen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Tripti Tamhane
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Janine Sophie Kemming
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Grigorii Nos
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Lasse Frank Voss
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Ulla Kring Hansen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Georg Michael Lauer
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Sine Reker Hadrup
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| |
Collapse
|
21
|
Nhu LNT, Chambers M, Chantratita N, Cheah PY, Day NP, Dejnirattisai W, Dunachie SJ, Grifoni A, Hamers RL, Hill J, Jones EY, Klenerman P, Mongkolsapaya J, Screaton G, Sette A, Stuart DI, Tan CW, Thwaites G, Thanh VD, Wang LF, Tan LV. Southeast Asia initiative to combat SARS-CoV-2 variants (SEACOVARIANTS) consortium. Wellcome Open Res 2024; 9:181. [PMID: 39022321 PMCID: PMC11252647 DOI: 10.12688/wellcomeopenres.20742.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 07/20/2024] Open
Abstract
A strong and effective COVID-19 and future pandemic responses rely on global efforts to carry out surveillance of infections and emerging SARS-CoV-2 variants and to act accordingly in real time. Many countries in Southeast Asia lack capacity to determine the potential threat of new variants, or other emerging infections. Funded by Wellcome, the Southeast Asia initiative to combat SARS-CoV-2 variants (SEACOVARIANTS) consortium aims to develop and apply a multidisciplinary research platform in Southeast Asia (SEA) for rapid assessment of the biological significance of SARS-CoV-2 variants, thereby informing coordinated local, regional and global responses to the COVID-19 pandemic. Our proposal is delivered by the Vietnam and Thailand Wellcome Africa Asia Programmes, bringing together a multidisciplinary team in Indonesia, Thailand and Vietnam with partners in Singapore, the UK and the USA. Herein we outline five work packages to deliver strengthened regional scientific capacity that can be rapidly deployed for future outbreak responses.
Collapse
Affiliation(s)
| | - Mary Chambers
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phaik Yeong Cheah
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nicholas P.J. Day
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanwisa Dejnirattisai
- Division of Emerging Infectious Disease, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Susanna J. Dunachie
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Alba Grifoni
- La Jolla Institute for Immunology, San Diego, California, USA
| | - Raph L. Hamers
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Oxford University Clinical Research Unit Indonesia, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Jennifer Hill
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - E. Yvonne Jones
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Paul Klenerman
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Juthathip Mongkolsapaya
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, England, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Gavin Screaton
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, England, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | | | - David I. Stuart
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Guy Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Vu Duy Thanh
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Le Van Tan
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
| | - SEACOVARIANTS Consortium
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Division of Emerging Infectious Disease, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- La Jolla Institute for Immunology, San Diego, California, USA
- Oxford University Clinical Research Unit Indonesia, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, England, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
22
|
Qian J, Zhang S, Wang F, Li J, Zhang J. What makes SARS-CoV-2 unique? Focusing on the spike protein. Cell Biol Int 2024; 48:404-430. [PMID: 38263600 DOI: 10.1002/cbin.12130] [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: 10/09/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
Severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) seriously threatens public health and safety. Genetic variants determine the expression of SARS-CoV-2 structural proteins, which are associated with enhanced transmissibility, enhanced virulence, and immune escape. Vaccination is encouraged as a public health intervention, and different types of vaccines are used worldwide. However, new variants continue to emerge, especially the Omicron complex, and the neutralizing antibody responses are diminished significantly. In this review, we outlined the uniqueness of SARS-CoV-2 from three perspectives. First, we described the detailed structure of the spike (S) protein, which is highly susceptible to mutations and contributes to the distinct infection cycle of the virus. Second, we systematically summarized the immunoglobulin G epitopes of SARS-CoV-2 and highlighted the central role of the nonconserved regions of the S protein in adaptive immune escape. Third, we provided an overview of the vaccines targeting the S protein and discussed the impact of the nonconserved regions on vaccine effectiveness. The characterization and identification of the structure and genomic organization of SARS-CoV-2 will help elucidate its mechanisms of viral mutation and infection and provide a basis for the selection of optimal treatments. The leaps in advancements regarding improved diagnosis, targeted vaccines and therapeutic remedies provide sound evidence showing that scientific understanding, research, and technology evolved at the pace of the pandemic.
Collapse
Affiliation(s)
- Jingbo Qian
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Shichang Zhang
- Department of Clinical Laboratory Medicine, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Fang Wang
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Jinming Li
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
- National Center for Clinical Laboratories, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing, China
| | - Jiexin Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| |
Collapse
|
23
|
Caldera F, Rolak S, Farraye FA, Necela BM, Cogen D, Zona EE, Schell TL, Ramirez OR, Almasry M, Chun K, Hayney MS, Knutson KL. Higher and Sustained Cell-Mediated Immune Responses After 3 Doses of mRNA COVID-19 Vaccine in Patients With Inflammatory Bowel Disease on Anti-Tumor Necrosis Factor Therapy. Clin Transl Gastroenterol 2024; 15:e00688. [PMID: 38349178 PMCID: PMC11042770 DOI: 10.14309/ctg.0000000000000688] [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: 08/31/2023] [Accepted: 01/19/2024] [Indexed: 04/26/2024] Open
Abstract
INTRODUCTION Studies suggest that the generation of durable T-cell immunity following coronavirus disease 2019 (COVID-19) vaccination protects against severe disease. The aim of this study was to measure cell-mediated immune response (CMIR) 1-2 months and 6 months after a third dose of a COVID-19 mRNA vaccine. METHODS This prospective study (HumoRal and CellULar initial and Sustained immunogenicity in patients with inflammatory bowel disease [IBD]) evaluated CMIR at 28-65 days (t 1 ) after dose 2, 28-65 days (t 2 ) (n = 183) and 6 months (±45 days) (t 3 ) (n = 167) after a third dose of an mRNA COVID-19 vaccine. A small cohort had blood sample available 28-65 days (t 4 ) (n = 55) after a fourth dose. Primary outcomes were CMIR at (t 2 ) and (t 3 ). Secondary outcomes included the effect of immunosuppressing IBD medications on CMIR and response at (t 4 ). RESULTS All patients had measurable CMIR at all time points. CMIR increased at t 2 compared with that at t 1 (median 1,467 responding cells per million (interquartile range [IQR] 410-5,971) vs 313 (94-960) P < 0.001). There was no significant waning in t 2 vs t 3 or significant boosting at t 4 . Those on anti-tumor necrosis factor monotherapy had a higher CMIR compared with those not on this therapy at t 2 (4,132 [IQR 1,136-8,795] vs 869 [IQR 343-3,221] P < 0.001) and t 3 (2,843 [IQR 596-6,459] vs 654 [IQR 143-2,067] P < 0.001). In univariable analysis, anti-tumor necrosis factor monotherapy was associated with a higher CMIR at t 2 ( P < 0.001) and t 3 ( P < 0.001) and confirmed in a multivariable model ( P < 0.001). DISCUSSION A third dose of a COVID-19 vaccine boosts CMIR, and the response is sustained in patients with IBD.
Collapse
Affiliation(s)
- Freddy Caldera
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, USA
| | - Stacey Rolak
- Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Francis A. Farraye
- Inflammatory Bowel Disease Center, Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Brian M. Necela
- Department of Immunology, Mayo Clinic, Jacksonville, Florida, USA
| | - Davitte Cogen
- Department of Immunology, Mayo Clinic, Jacksonville, Florida, USA
| | - Emily E. Zona
- University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Trevor L. Schell
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Oscar Ramirez Ramirez
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Mazen Almasry
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kelly Chun
- LabCorp, R&D and Specialty Medicine, Calabasas, CA, USA
| | - Mary S. Hayney
- School of Pharmacy, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, USA
| | - Keith L. Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
24
|
Halvorson T, Ivison S, Huang Q, Ladua G, Yotis DM, Mannar D, Subramaniam S, Ferreira VH, Kumar D, Belga S, Levings MK. SARS-CoV-2 Variants Omicron BA.4/5 and XBB.1.5 Significantly Escape T Cell Recognition in Solid-organ Transplant Recipients Vaccinated Against the Ancestral Strain. Transplantation 2024; 108:e49-e62. [PMID: 38012843 DOI: 10.1097/tp.0000000000004873] [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] [Indexed: 11/29/2023]
Abstract
BACKGROUND Immune-suppressed solid-organ transplant recipients (SOTRs) display impaired humoral responses to COVID-19 vaccination, but T cell responses are incompletely understood. SARS-CoV-2 variants Omicron BA.4/5 (BA.4/5) and XBB.1.5 escape neutralization by antibodies induced by vaccination or infection with earlier strains, but T cell recognition of these lineages in SOTRs is unclear. METHODS We characterized Spike-specific T cell responses to ancestral SARS-CoV-2 and BA.4/5 peptides in 42 kidney, liver, and lung transplant recipients throughout a 3- or 4-dose ancestral Spike mRNA vaccination schedule. As the XBB.1.5 variant emerged during the study, we tested vaccine-induced T cell responses in 10 additional participants using recombinant XBB.1.5 Spike protein. Using an optimized activation-induced marker assay, we quantified circulating Spike-specific CD4 + and CD8 + T cells based on antigen-stimulated expression of CD134, CD69, CD25, CD137, and/or CD107a. RESULTS Vaccination strongly induced SARS-CoV-2-specific T cells, including BA.4/5- and XBB.1.5-reactive T cells, which remained detectable over time and further increased following a fourth dose. However, responses to BA.4/5 (1.34- to 1.67-fold lower) XBB.1.5 (2.0- to 18-fold lower) were significantly reduced in magnitude compared with ancestral strain responses. CD4 + responses correlated with anti-receptor-binding domain antibodies and predicted subsequent antibody responses in seronegative individuals. Lung transplant recipients receiving prednisone and older adults displayed weaker responses. CONCLUSIONS Ancestral strain vaccination stimulates BA.4/5 and XBB.1.5-cross-reactive T cells in SOTRs, but at lower magnitudes. Antigen-specific T cells can predict future antibody responses. Our data support monitoring both humoral and cellular immunity in SOTRs to track COVID-19 vaccine immunogenicity against emerging variants.
Collapse
Affiliation(s)
- Torin Halvorson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Sabine Ivison
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Qing Huang
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Gale Ladua
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Infection and Immunity Research Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Demitra M Yotis
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Dhiraj Mannar
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sriram Subramaniam
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Victor H Ferreira
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Deepali Kumar
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Sara Belga
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Infection and Immunity Research Centre, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
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
|
26
|
Ogando-Rivas E, Castillo P, Yang C, Trivedi V, Zhang D, Pohl-Guimarães F, Liu R, Barpujari A, Candelario KM, Mendez-Gomez H, Sayour EJ, Mitchell DA. Expanded specific T cells to hypomutated regions of the SARS-CoV-2 using mRNA electroporated antigen-presenting cells. Mol Ther Methods Clin Dev 2024; 32:101192. [PMID: 38327807 PMCID: PMC10847775 DOI: 10.1016/j.omtm.2024.101192] [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: 04/10/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024]
Abstract
The COVID-19 pandemic has caused about seven million deaths worldwide. Preventative vaccines have been developed including Spike gp mRNA-based vaccines that provide protection to immunocompetent patients. However, patients with primary immunodeficiencies, patients with cancer, or hematopoietic stem cell transplant recipients are not able to mount robust immune responses against current vaccine approaches. We propose to target structural SARS-CoV-2 antigens (i.e., Spike gp, Membrane, Nucleocapsid, and Envelope) using circulating human antigen-presenting cells electroporated with full length SARS-CoV-2 structural protein-encoding mRNAs to activate and expand specific T cells. Based on the Th1-type cytokine and cytolytic enzyme secretion upon antigen rechallenge, we were able to generate SARS-CoV-2 specific T cells in up to 70% of unexposed unvaccinated healthy donors (HDs) after 3 subsequent stimulations and in 100% of recovered patients (RPs) after 2 stimulations. By means of SARS-CoV-2 specific TCRβ repertoire analysis, T cells specific to Spike gp-derived hypomutated regions were identified in HDs and RPs despite viral genomic evolution. Hence, we demonstrated that SARS-CoV-2 mRNA-loaded antigen-presenting cells are effective activating and expanding COVID19-specific T cells. This approach represents an alternative to patients who are not able to mount adaptive immune responses to current COVID-19 vaccines with potential protection across new variants that have conserved genetic regions.
Collapse
Affiliation(s)
- Elizabeth Ogando-Rivas
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Paul Castillo
- UF Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Changlin Yang
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Vrunda Trivedi
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Dingpeng Zhang
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Fernanda Pohl-Guimarães
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Ruixuan Liu
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Arnav Barpujari
- UF Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Kate M. Candelario
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Hector Mendez-Gomez
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Elias J. Sayour
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- UF Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Duane A. Mitchell
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| |
Collapse
|
27
|
Ho CL, Yen LC, Huang HW, Lu CC, Hung YJ, Liao CL, Hung CM, Chiu KC. Long-term analysis of humoral responses and spike-specific T cell memory to Omicron variants after different COVID-19 vaccine regimens. Front Immunol 2024; 15:1340645. [PMID: 38533494 PMCID: PMC10963495 DOI: 10.3389/fimmu.2024.1340645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
Background The emergence of SARS-CoV-2 variants has raised concerns about the sustainability of vaccine-induced immunity. Little is known about the long-term humoral responses and spike-specific T cell memory to Omicron variants, with specific attention to BA.4/5, BQ.1.1, and XBB.1. Methods We assessed immune responses in 50 uninfected individuals who received varying three-dose vaccination combinations (2X AstraZeneca + 1X Moderna, 1X AstraZeneca + 2X Moderna, and 3X Moderna) against wild-type (WT) and Omicron variants at eight months post-vaccination. The serum antibody titers were analyzed by enzyme-linked immunosorbent assays (ELISA), and neutralizing activities were examined by pseudovirus and infectious SARS-CoV-2 neutralization assays. T cell reactivities and their memory phenotypes were determined by flow cytometry. Results We found that RBD-specific antibody titers, neutralizing activities, and CD4+ T cell reactivities were reduced against Omicron variants compared to WT. In contrast, CD8+ T cell responses, central memory, effector memory, and CD45RA+ effector memory T cells remained unaffected upon stimulation with the Omicron peptide pool. Notably, CD4+ effector memory T cells even exhibited a higher proportion of reactivity against Omicron variants. Furthermore, participants who received three doses of the Moderna showed a more robust response regarding neutralization and CD8+ T cell reactions than other three-dose vaccination groups. Conclusion Reduction of humoral and CD4+ T cell responses against Omicron variants in vaccinees suggested that vaccine effectiveness after eight months may not have sufficient protection against the new emerging variants, which provides valuable information for future vaccination strategies such as receiving BA.4/5 or XBB.1-based bivalent vaccines.
Collapse
Affiliation(s)
- Chia-Lo Ho
- Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Li-Chen Yen
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Hong-Wei Huang
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Chun-Chi Lu
- Division of Rheumatology/Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Jen Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Len Liao
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chin-Mao Hung
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Chou Chiu
- Division of General Dentistry, Taichung Armed Forces General Hospital, Taichung, Taiwan
- School of Dentistry, China Medical University, Taichung, Taiwan
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
28
|
Bricio-Moreno L, Barreto de Albuquerque J, Neary JM, Nguyen T, Kuhn LF, Yeung Y, Hastie KM, Landeras-Bueno S, Olmedillas E, Hariharan C, Nathan A, Getz MA, Gayton AC, Khatri A, Gaiha GD, Ollmann Saphire E, Luster AD, Moon JJ. Identification of mouse CD4 + T cell epitopes in SARS-CoV-2 BA.1 spike and nucleocapsid for use in peptide:MHCII tetramers. Front Immunol 2024; 15:1329846. [PMID: 38529279 PMCID: PMC10961420 DOI: 10.3389/fimmu.2024.1329846] [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: 10/30/2023] [Accepted: 01/29/2024] [Indexed: 03/27/2024] Open
Abstract
Understanding adaptive immunity against SARS-CoV-2 is a major requisite for the development of effective vaccines and treatments for COVID-19. CD4+ T cells play an integral role in this process primarily by generating antiviral cytokines and providing help to antibody-producing B cells. To empower detailed studies of SARS-CoV-2-specific CD4+ T cell responses in mouse models, we comprehensively mapped I-Ab-restricted epitopes for the spike and nucleocapsid proteins of the BA.1 variant of concern via IFNγ ELISpot assay. This was followed by the generation of corresponding peptide:MHCII tetramer reagents to directly stain epitope-specific T cells. Using this rigorous validation strategy, we identified 6 immunogenic epitopes in spike and 3 in nucleocapsid, all of which are conserved in the ancestral Wuhan strain. We also validated a previously identified epitope from Wuhan that is absent in BA.1. These epitopes and tetramers will be invaluable tools for SARS-CoV-2 antigen-specific CD4+ T cell studies in mice.
Collapse
Affiliation(s)
- Laura Bricio-Moreno
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Juliana Barreto de Albuquerque
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Jake M. Neary
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
| | - Thao Nguyen
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
| | - Lucy F. Kuhn
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
| | - YeePui Yeung
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
| | - Kathryn M. Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Sara Landeras-Bueno
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Eduardo Olmedillas
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Chitra Hariharan
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Anusha Nathan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA, United States
| | - Matthew A. Getz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Alton C. Gayton
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Ashok Khatri
- Harvard Medical School, Boston, MA, United States
- Endocrine Division, MGH, Boston, MA, United States
| | - Gaurav D. Gaiha
- Harvard Medical School, Boston, MA, United States
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Division of Gastroenterology, MGH, Boston, MA, United States
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Andrew D. Luster
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - James J. Moon
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Pulmonary and Critical Care Medicine, MGH, Boston, MA, United States
| |
Collapse
|
29
|
Vasconcelos LDCM, Leony LM, Camelier AA, Meireles AC, Oliveira Júnior ALFD, Bandeira AC, Macedo YSF, Duarte AO, Van Voorhis W, Siqueira ICD, Santos FLN. Usefulness of receptor binding domain protein-based serodiagnosis of COVID-19. IJID REGIONS 2024; 10:1-8. [PMID: 38045864 PMCID: PMC10687696 DOI: 10.1016/j.ijregi.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/31/2023] [Accepted: 11/05/2023] [Indexed: 12/05/2023]
Abstract
Objectives This study evaluated the performance of recombinant receptor binding domain (RBD) protein-based enzyme-linked immunosorbent assays (RBD-ELISAs) for detecting anti-SARS-CoV-2 immunoglobulin (Ig) G and IgM antibodies. Methods In this study, 705 sera from SARS-CoV-2-infected individuals and 315 sera from healthy individuals were analyzed. Results The RBD-ELISA IgG exhibited high specificity (99.1%) and moderate sensitivity (48.0%), with an overall diagnostic accuracy of 73.5%. RBD-ELISA IgM demonstrated specificity at 94.6% and sensitivity at 51.1%, with an accuracy of 72.8%. Both assays displayed improved performance when analyzing samples collected 15-21 days post-symptom onset, achieving sensitivity and accuracy exceeding 88% and 90%, respectively. Combining RBD-ELISA IgG and IgM in parallel analysis enhanced sensitivity to 98.6% and accuracy to 96.2%. Comparing these RBD-ELISAs with commercially available tests, the study found overlapping sensitivity and similar specificity values. Notably, the combined RBD-ELISA IgG and IgM showed superior performance. Cross-reactivity analysis revealed low false-positive rates (4.4% for IgG, 3.7% for IgM), primarily with viral infections. Conclusion This research underscores the potential of RBD-based ELISAs for COVID-19 diagnosis, especially when assessing samples collected 15-21 days post-symptom onset and utilizing a parallel testing approach. The RBD protein's immunogenicity and specificity make it a valuable tool for serodiagnosis, offering an alternative to polymerase chain reaction-based methods, particularly in resource-limited settings.
Collapse
Affiliation(s)
| | - Leonardo Maia Leony
- Advanced Public Health Laboratory, Gonçalo Moniz Institute (IGM), Oswaldo Cruz Foundation (FIOCRUZ-RJ), Salvador, Brazil
| | - Aquiles Assunção Camelier
- Aliança D'Or Hospital, Salvador, Brazil
- Bahia School of Medicine and Public Health, Salvador, Brazil
- State University of Bahia, Salvador, Brazil
| | | | | | | | - Yasmin Santos Freitas Macedo
- Laboratory of Experimental Pathology, Institute Gonçalo Moniz, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
| | - Alan Oliveira Duarte
- Laboratory of Experimental Pathology, Institute Gonçalo Moniz, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
| | | | - Isadora Cristina de Siqueira
- Laboratory of Experimental Pathology, Institute Gonçalo Moniz, Oswaldo Cruz Foundation (FIOCRUZ-BA), Salvador, Brazil
- Integrated Translational Program in Chagas Disease from FIOCRUZ (Fio-Chagas), Oswaldo Cruz Foundation (FIOCRUZ-RJ), Rio de Janeiro, Brazil
| | - Fred Luciano Neves Santos
- Advanced Public Health Laboratory, Gonçalo Moniz Institute (IGM), Oswaldo Cruz Foundation (FIOCRUZ-RJ), Salvador, Brazil
- Integrated Translational Program in Chagas Disease from FIOCRUZ (Fio-Chagas), Oswaldo Cruz Foundation (FIOCRUZ-RJ), Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Ceolin C, De Rui M, Simonato C, Vergadoro M, Cazzavillan S, Acunto V, Papa MV, Trapella GS, Zanforlini BM, Curreri C, Bertocco A, Devita M, Coin A, Sergi G. Sarcopenic patients "get even": The impact of COVID-19 vaccination on mortality. Exp Gerontol 2024; 187:112382. [PMID: 38369251 DOI: 10.1016/j.exger.2024.112382] [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: 01/16/2024] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Coronavirus Disease-2019 (COVID-19), driven by the SARS-CoV-2 virus, has disproportionately affected the elderly, with comorbidities like sarcopenia worsening prognosis. Considering the significant impact of RNA vaccines on survival rates in this population, our objective is to investigate the impact of vaccination on the survival of hospitalized elderly patients with COVID-19, considering the presence or absence of sarcopenia. METHODS Prospective study conducted on 159 patients aged>65 years from September 2021 to March 2022. Data about clinical and body composition, and mortality at 12-months after discharge were recorded. Sarcopenia was diagnosed according to the 2019 European Consensus criteria. RESULTS At the twelfth month post-discharge, vaccinated sarcopenic individuals exhibited a mortality risk similar to vaccinated non-sarcopenic individuals, and lower than unvaccinated non-sarcopenic patients. Cox regression analysis, adjusted for age, gender, comorbidity, functional and vaccinal status, showed that the presence of sarcopenia did not significantly impact the risk of death within 12-months post-discharge. DISCUSSION Vaccination emerges as a protective measure for sarcopenic patients, countering the potential adverse effects of sarcopenia on COVID-19 outcomes, underscoring the importance of immunization in the frail elderly with a call for meticulous monitoring of its benefits. CONCLUSIONS Our study represents the first attempt to analyze the vaccine's effect on survival in sarcopenic hospitalized older adults with COVID-19. The administration of vaccination to sarcopenic patients proves pivotal, as its omission could lead to notably unfavorable outcomes within this specific population.
Collapse
Affiliation(s)
- Chiara Ceolin
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy.
| | - Marina De Rui
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | - Cristina Simonato
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | - Margherita Vergadoro
- Department of Medicine (DIMED), Department of Women's and Children's Health, University of Padua, Italy
| | - Sara Cazzavillan
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | - Vittorio Acunto
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | - Mario Virgilio Papa
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | | | | | - Chiara Curreri
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | - Anna Bertocco
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | - Maria Devita
- Department of General Psychology (DPG), University of Padua, Italy
| | - Alessandra Coin
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| | - Giuseppe Sergi
- Department of Medicine (DIMED), Geriatrics Division, University of Padua, Italy
| |
Collapse
|
31
|
Maino A, Amen A, Plumas J, Bouquet L, Deschamps M, Saas P, Chaperot L, Manches O. Development of a New Off-the-Shelf Plasmacytoid Dendritic Cell-Based Approach for the Expansion and Characterization of SARS-CoV-2-Specific T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:825-833. [PMID: 38214610 DOI: 10.4049/jimmunol.2300704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/20/2023] [Indexed: 01/13/2024]
Abstract
Global vaccination against COVID-19 has been widely successful; however, there is a need for complementary immunotherapies in severe forms of the disease and in immunocompromised patients. Cytotoxic CD8+ T cells have a crucial role in disease control, but their function can be dysregulated in severe forms of the disease. We report here a cell-based approach using a plasmacytoid dendritic cell line (PDC*line) to expand in vitro specific CD8+ responses against COVID-19 Ags. We tested the immunogenicity of eight HLA-A*02:01 restricted peptides derived from diverse SARS-Cov-2 proteins, selected by bioinformatics analyses in unexposed and convalescent donors. Higher ex vivo frequencies of specific T cells against these peptides were found in convalescent donors compared with unexposed donors, suggesting in situ T cell expansion upon viral infection. The peptide-loaded PDC*line induced robust CD8+ responses with total amplification rates that led up to a 198-fold increase in peptide-specific CD8+ T cell frequencies for a single donor. Of note, six of eight selected peptides provided significant amplifications, all of which were conserved between SARS-CoV variants and derived from the membrane, the spike protein, the nucleoprotein, and the ORF1ab. Amplified and cloned antiviral CD8+ T cells secreted IFN-γ upon peptide-specific activation. Furthermore, specific TCR sequences were identified for two highly immunogenic Ags. Hence, PDC*line represents an efficient platform to identify immunogenic viral targets for future immunotherapies.
Collapse
Affiliation(s)
- Anthony Maino
- Etablissement Français du Sang, Recherche et Développement, Grenoble, France
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Axelle Amen
- Laboratoire d'Immunologie, Centre Hospitalier Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, CNRS, CEA, UMR 5075, Institut de Biologie Structurale, Grenoble, France
| | - Joël Plumas
- Etablissement Français du Sang, Recherche et Développement, Grenoble, France
- PDC*line Pharma SAS, Grenoble, France
| | - Lucie Bouquet
- Université de Franche-Comté, Etablissement Français du Sang, INSERM, UMR RIGHT, Besançon, France
| | - Marina Deschamps
- Université de Franche-Comté, Etablissement Français du Sang, INSERM, UMR RIGHT, Besançon, France
| | - Philippe Saas
- Etablissement Français du Sang, Recherche et Développement, Grenoble, France
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence Chaperot
- Etablissement Français du Sang, Recherche et Développement, Grenoble, France
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Olivier Manches
- Etablissement Français du Sang, Recherche et Développement, Grenoble, France
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
32
|
Gerhards C, Steingass M, Heininger A, Lange B, Hetjens M, Gerigk M, Neumaier M, Evliyaoglu O, Kittel M. The Impact of Clinical Factors and SARS-CoV-2 Variants on Antibody Production in Vaccinated German Healthcare Professionals Infected Either with the Delta or the Omicron Variant. Vaccines (Basel) 2024; 12:163. [PMID: 38400146 PMCID: PMC10893335 DOI: 10.3390/vaccines12020163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The aim of the rapid introduction of vaccines during the COVID-19 pandemic was a reduction in SARS-CoV-2 transmission and a less frequent occurrence of severe COVID-19 courses. Thus, we evaluated COVID-19 severity in vaccinated individuals to examine variant-specific symptom characteristics and their clinical impact on the serological immune response. METHODS A total of 185 individuals previously vaccinated against and infected with the SARS-CoV-2 Delta (B.1.617.2) or Omicron (BA.4 and BA.5) variant, were enrolled for anti-SARS-CoV-2 anti-N- and anti-RBD/S1-Ig level detection. A structured survey regarding medical history was conducted. RESULTS In 99.5 percent of cases, outpatient treatment was satisfactory. Specific symptoms associated with variants included ageusia and anosmia in patients with Delta infections and throat pain in Omicron infections. Among Delta-infected individuals with specific symptoms, significantly higher levels of anti-N antibodies were observed. CONCLUSION Our study identified variant-specific differences in the amount of SARS-CoV-2 antibody production and COVID-19 symptoms. Despite this, vaccinated individuals with Omicron or Delta infections generally experienced mild disease courses. Additionally, asymptomatic individuals exhibit lower anti-SARS-CoV-2 antibody levels, indicating a clinical correlation between disease-specific antibodies and distinct symptoms, particularly in the case of the Delta variant. In follow-up studies, exploring post-COVID syndrome and focusing on cognitive symptoms in the acute phase of Omicron infections is crucial as it has the potential to longitudinally impact the lives of those affected.
Collapse
Affiliation(s)
- Catharina Gerhards
- Institute for Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Marlene Steingass
- Institute for Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Alexandra Heininger
- Department of Hygiene, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Bettina Lange
- Department of Hygiene, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Michael Hetjens
- Department of Biomedical Informatics, Center for Preventive Medicine and Digital Health Baden-Württemberg, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg of University, 68167 Mannheim, Germany
| | - Marlis Gerigk
- Institute of Medical Microbiology and Hygiene, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Michael Neumaier
- Institute for Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Osman Evliyaoglu
- Institute for Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Maximilian Kittel
- Institute for Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
33
|
Zhan H, Xie Y, Liu Y, Cheng L, Xu Y, Qu X, Li C, Guo X, Li H, Wang Y, Dai E, Wang L, Gao H, Li Y. Omicron BA.4/5 neutralization and cell-mediated immune responses in relation to baseline immune status and breakthrough infection among PLWH: A follow-up cohort study. J Med Virol 2024; 96:e29446. [PMID: 38345110 DOI: 10.1002/jmv.29446] [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: 10/25/2023] [Revised: 01/04/2024] [Accepted: 01/21/2024] [Indexed: 02/15/2024]
Abstract
There is a paucity of data on hybrid immunity (vaccination plus breakthrough infection [BI]), especially cell-mediated responses to Omicron among immunosuppressed patients. We aim to investigate humoral and cellular responses to Omicron BA.4/5 among people living with HIV (PLWH) with/without BIs, the most prevalent variant of concern after the reopening of China. Based on our previous study, we enrolled 77 PLWH with baseline immune status of severe acute respiratory syndrome coronavirus 2 specific antibodies after inactivated vaccination. "Correlates of protection," including serological immunoassays, T cell phenotypes and memory B cells (MBC) were determined in PLWH without and with BI, together with 16 PLWH with reinfections. Higher inhibition rate of neutralizing antibodies (NAb) against BA.4/5 was elicited among PLWH with BI than those without. Omicron-reactive IL4+ CD8+ T cells were significantly elevated in PLWH experienced postvaccine infection contrasting with those did not. NAb towards wild type at baseline was associated with prolonged negative conversion time for PLWH whereas intermediate MBCs serve as protecting effectors. We uncovered that hybrid immunity intensified more protection on BA.4/5 than vaccination did. Strengthened surveillance on immunological parameters and timely clinical intervention on PLWH deficient in protection would reduce the severity and mortality in the context of coexistence with new Omicron subvariants.
Collapse
Affiliation(s)
- Haoting Zhan
- Department of Clinical Laboratory, State key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuchen Xie
- School of Public Health, North China University of Science and Technology, Tangshan, China
- Department of Laboratory Medicine, Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Yongmei Liu
- Department of Clinical Laboratory, State key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linlin Cheng
- Department of Clinical Laboratory, State key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yi Xu
- Department of Laboratory Medicine, Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Xiaojing Qu
- Department of AIDS, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Chen Li
- Department of AIDS, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Xinru Guo
- School of Public Health, North China University of Science and Technology, Tangshan, China
- Department of Laboratory Medicine, Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Haolong Li
- Department of Clinical Laboratory, State key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuling Wang
- Department of AIDS, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Erhei Dai
- Department of Laboratory Medicine, Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Lijing Wang
- Department of AIDS, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Huixia Gao
- Department of Laboratory Medicine, Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, North China University of Science and Technology, Tangshan, China
| | - Yongzhe Li
- Department of Clinical Laboratory, State key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Gan M, Cao J, Zhang Y, Fu H, Lin X, Ouyang Q, Xu X, Yuan Y, Fan X. Landscape of T cell epitopes displays hot mutations of SARS-CoV-2 variant spikes evading cellular immunity. J Med Virol 2024; 96:e29452. [PMID: 38314852 DOI: 10.1002/jmv.29452] [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: 09/14/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
The continuous evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been accompanied by the emergence of viral mutations that pose a great challenge to existing vaccine strategies. It is not fully understood with regard to the role of mutations on the SARS-CoV-2 spike protein from emerging viral variants in T cell immunity. In the current study, recombinant eukaryotic plasmids were constructed as DNA vaccines to express the spike protein from multiple SARS-CoV-2 strains. These DNA vaccines were used to immunize BALB/c mice, and cross-T cell responses to the spike protein from these viral strains were quantitated using interferon-γ (IFN-γ) Elispot. Peptides covering the full-length spike protein from different viral strains were used to detect epitope-specific IFN-γ+ CD4+ and CD8+ T cell responses by fluorescence-activated cell sorting. SARS-CoV-2 Delta and Omicron BA.1 strains were found to have broad T cell cross-reactivity, followed by the Beta strain. The landscapes of T cell epitopes on the spike protein demonstrated that at least 30 mutations emerging from Alpha to Omicron BA.5 can mediate the escape of T cell immunity. Omicron and its sublineages have 19 out of these 30 mutations, most of which are new, and a few are inherited from ancient circulating variants of concerns. The cross-T cell immunity between SARS-CoV-2 prototype strain and Omicron strains can be attributed to the T cell epitopes located in the N-terminal domain (181-246 aa [amino acids], 271-318 aa) and C-terminal domain (1171-1273 aa) of the spike protein. These findings provide in vivo evidence for optimizing vaccine manufacturing and immunization strategies for current or future viral variants.
Collapse
Affiliation(s)
- Mengze Gan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jinge Cao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yandi Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Fu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaosong Lin
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Ouyang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyue Xu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Yuan
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xionglin Fan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Kim SH, Kim J, Jung S, Noh JY, Kim J, Park H, Song YG, Peck KR, Park SH, Park MS, Ko JH, Song JY, Choi JY, Jung MK, Shin EC. Omicron BA.2 breakthrough infection elicits CD8 + T cell responses recognizing the spike of later Omicron subvariants. Sci Immunol 2024; 9:eade6132. [PMID: 38241400 DOI: 10.1126/sciimmunol.ade6132] [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: 08/27/2022] [Accepted: 12/14/2023] [Indexed: 01/21/2024]
Abstract
Here, we examine peripheral blood memory T cell responses against the SARS-CoV-2 BA.4/BA.5 variant spike among vaccinated individuals with or without Omicron breakthrough infections. We provide evidence supporting a lack of original antigenic sin in CD8+ T cell responses targeting the spike. We show that BNT162b2-induced memory T cells respond to the BA.4/BA.5 spike. Among individuals with BA.1/BA.2 breakthrough infections, IFN-γ-producing CD8+ T cell responses against the BA.4/BA.5 spike increased. In a subgroup with BA.2 breakthrough infections, IFN-γ-producing CD8+ T cell responses against the BA.2-mutated spike region increased and correlated directly with responses against the BA.4/BA.5 spike, indicating that BA.2 spike-specific CD8+ T cells elicited by BA.2 breakthrough infection cross-react with the BA.4/BA.5 spike. We identified CD8+ T cell epitope peptides that are present in the spike of BA.2 and BA.4/BA.5 but not the original spike. These peptides are fully conserved in the spike of now-dominant XBB lineages. Our study shows that breakthrough infection by early Omicron subvariants elicits CD8+ T cell responses that recognize epitopes within the spike of newly emerging subvariants.
Collapse
Affiliation(s)
- Sang-Hoon Kim
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Jihye Kim
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Sungmin Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Jinnam Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heedo Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Young Goo Song
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Jun Yong Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Kyung Jung
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Eui-Cheol Shin
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
36
|
Guimaraes LC, Costa PAC, Scalzo Júnior SRA, Ferreira HAS, Braga ACS, de Oliveira LC, Figueiredo MM, Shepherd S, Hamilton A, Queiroz-Junior CM, da Silva WN, da Silva NJA, Rodrigues Alves MT, Santos AK, de Faria KKS, Marim FM, Fukumasu H, Birbrair A, Teixeira-Carvalho A, de Aguiar RS, Mitchell MJ, Teixeira MM, Vasconcelos Costa V, Frezard F, Guimaraes PPG. Nanoparticle-based DNA vaccine protects against SARS-CoV-2 variants in female preclinical models. Nat Commun 2024; 15:590. [PMID: 38238326 PMCID: PMC10796936 DOI: 10.1038/s41467-024-44830-1] [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: 03/31/2023] [Accepted: 01/05/2024] [Indexed: 01/22/2024] Open
Abstract
A safe and effective vaccine with long-term protection against SARS-CoV-2 variants of concern (VOCs) is a global health priority. Here, we develop lipid nanoparticles (LNPs) to provide safe and effective delivery of plasmid DNA (pDNA) and show protection against VOCs in female small animal models. Using a library of LNPs encapsulating unique barcoded DNA (b-DNA), we screen for b-DNA delivery after intramuscular administration. The top-performing LNPs are further tested for their capacity of pDNA uptake in antigen-presenting cells in vitro. The lead LNP is used to encapsulate pDNA encoding the HexaPro version of SARS-CoV-2 spike (LNP-HPS) and immunogenicity and protection is tested in vivo. LNP-HPS elicit a robust protective effect against SARS-CoV-2 Gamma (P.1), correlating with reduced lethality, decreased viral load in the lungs and reduced lung damage. LNP-HPS induce potent humoral and T cell responses against P.1, and generate high levels of neutralizing antibodies against P.1 and Omicron (B.1.1.529). Our findings indicate that the protective efficacy and immunogenicity elicited by LNP-HPS are comparable to those achieved by the approved COVID-19 vaccine from Biontech/Pfizer in animal models. Together, these findings suggest that LNP-HPS hold great promise as a vaccine candidate against VOCs.
Collapse
Affiliation(s)
- Lays Cordeiro Guimaraes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Pedro Augusto Carvalho Costa
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Sérgio Ricardo Aluotto Scalzo Júnior
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Heloísa Athaydes Seabra Ferreira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Ana Carolina Soares Braga
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Leonardo Camilo de Oliveira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | | | - Sarah Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Alex Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | | | - Walison Nunes da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Natália Jordana Alves da Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Marco Túllio Rodrigues Alves
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Anderson Kenedy Santos
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Kevin Kelton Santos de Faria
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Fernanda Martins Marim
- Department of Genetics, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Heidge Fukumasu
- Department of Animal Science and Food Engineering, University of São Paulo, Pirassununga, 13635-900, São Paulo, Brazil
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Madison, 53706, WI, USA
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, 30190-009, Minas Gerais, Brazil
| | - Renato Santana de Aguiar
- Department of Genetics, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Frederic Frezard
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil
| | - Pedro Pires Goulart Guimaraes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Minas Gerais, Brazil.
| |
Collapse
|
37
|
Gainullin M, Federico L, Røkke Osen J, Chaban V, Kared H, Alirezaylavasani A, Lund-Johansen F, Wildendahl G, Jacobsen JA, Sarwar Anjum H, Stratford R, Tennøe S, Malone B, Clancy T, Vaage JT, Henriksen K, Wüsthoff L, Munthe LA. People who use drugs show no increase in pre-existing T-cell cross-reactivity toward SARS-CoV-2 but develop a normal polyfunctional T-cell response after standard mRNA vaccination. Front Immunol 2024; 14:1235210. [PMID: 38299149 PMCID: PMC10827924 DOI: 10.3389/fimmu.2023.1235210] [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: 06/05/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
People who use drugs (PWUD) are at a high risk of contracting and developing severe coronavirus disease 2019 (COVID-19) and other infectious diseases due to their lifestyle, comorbidities, and the detrimental effects of opioids on cellular immunity. However, there is limited research on vaccine responses in PWUD, particularly regarding the role that T cells play in the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we show that before vaccination, PWUD did not exhibit an increased frequency of preexisting cross-reactive T cells to SARS-CoV-2 and that, despite the inhibitory effects that opioids have on T-cell immunity, standard vaccination can elicit robust polyfunctional CD4+ and CD8+ T-cell responses that were similar to those found in controls. Our findings indicate that vaccination stimulates an effective immune response in PWUD and highlight targeted vaccination as an essential public health instrument for the control of COVID-19 and other infectious diseases in this group of high-risk patients.
Collapse
Affiliation(s)
- Murat Gainullin
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NEC OncoImmunity AS, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Lorenzo Federico
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Julie Røkke Osen
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Viktoriia Chaban
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Hassen Kared
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Amin Alirezaylavasani
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | - John T. Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kathleen Henriksen
- Agency for Social and Welfare Services, Oslo, Norway
- Student Health Services, University of Oslo, Oslo, Norway
| | - Linda Wüsthoff
- Unit for Clinical Research on Addictions, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Addiction Reasearch, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A. Munthe
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
38
|
Vecchio E, Rotundo S, Veneziano C, Abatino A, Aversa I, Gallo R, Giordano C, Serapide F, Fusco P, Viglietto G, Cuda G, Costanzo F, Russo A, Trecarichi EM, Torti C, Palmieri C. The spike-specific TCRβ repertoire shows distinct features in unvaccinated or vaccinated patients with SARS-CoV-2 infection. J Transl Med 2024; 22:33. [PMID: 38185632 PMCID: PMC10771664 DOI: 10.1186/s12967-024-04852-1] [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: 10/22/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND The evolving variants of SARS-CoV-2 may escape immunity from prior infections or vaccinations. It's vital to understand how immunity adapts to these changes. Both infection and mRNA vaccination induce T cells that target the Spike protein. These T cells can recognize multiple variants, such as Delta and Omicron, even if neutralizing antibodies are weakened. However, the degree of recognition can vary among people, affecting vaccine efficacy. Previous studies demonstrated the capability of T-cell receptor (TCR) repertoire analysis to identify conserved and immunodominant peptides with cross-reactive potential among variant of concerns. However, there is a need to extend the analysis of the TCR repertoire to different clinical scenarios. The aim of this study was to examine the Spike-specific TCR repertoire profiles in natural infections and those with combined natural and vaccine immunity. METHODS A T-cell enrichment approach and bioinformatic tools were used to investigate the Spike-specific TCRβ repertoire in peripheral blood mononuclear cells of previously vaccinated (n = 8) or unvaccinated (n = 6) COVID-19 patients. RESULTS Diversity and clonality of the TCRβ repertoire showed no significant differences between vaccinated and unvaccinated groups. When comparing the TCRβ data to public databases, 692 unique TCRβ sequences linked to S epitopes were found in the vaccinated group and 670 in the unvaccinated group. TCRβ clonotypes related to spike regions S135-177, S264-276, S319-350, and S448-472 appear notably more prevalent in the vaccinated group. In contrast, the S673-699 epitope, believed to have super antigenic properties, is observed more frequently in the unvaccinated group. In-silico analyses suggest that mutations in epitopes, relative to the main SARS-CoV-2 variants of concern, don't hinder their cross-reactive recognition by associated TCRβ clonotypes. CONCLUSIONS Our findings reveal distinct TCRβ signatures in vaccinated and unvaccinated individuals with COVID-19. These differences might be associated with disease severity and could influence clinical outcomes. TRIAL REGISTRATION FESR/FSE 2014-2020 DDRC n. 585, Action 10.5.12, noCOVID19@UMG.
Collapse
Affiliation(s)
- Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
- Interdepartmental Centre of Services, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Salvatore Rotundo
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Claudia Veneziano
- Interdepartmental Centre of Services, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Antonio Abatino
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Ilenia Aversa
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Raffaella Gallo
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Caterina Giordano
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Francesca Serapide
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Paolo Fusco
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
| | - Francesco Costanzo
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy
- Interdepartmental Centre of Services, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Alessandro Russo
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Enrico Maria Trecarichi
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Carlo Torti
- Department of Medical and Surgical Sciences, Chair of Infectious and Tropical Diseases, University "Magna Graecia", 88100, Catanzaro, Italy
| | - Camillo Palmieri
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Viale Europa, 88100, Catanzaro, Italy.
| |
Collapse
|
39
|
Prins MLM, Roozen GVT, Pothast CR, Huisman W, van Binnendijk R, den Hartog G, Kuiper VP, Prins C, Janse JJ, Lamers OAC, Koopman JPR, Kruithof AC, Kamerling IMC, Dijkland RC, de Kroon AC, Azimi S, Feltkamp MCW, Kuijer M, Jochems SP, Heemskerk MHM, Rosendaal FR, Roestenberg M, Visser LG, Roukens AHE. Immunogenicity and reactogenicity of intradermal mRNA-1273 SARS-CoV-2 vaccination: a non-inferiority, randomized-controlled trial. NPJ Vaccines 2024; 9:1. [PMID: 38167735 PMCID: PMC10761693 DOI: 10.1038/s41541-023-00785-w] [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: 09/12/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024] Open
Abstract
Fractional dosing can be a cost-effective vaccination strategy to accelerate individual and herd immunity in a pandemic. We assessed the immunogenicity and safety of primary intradermal (ID) vaccination, with a 1/5th dose compared with the standard intramuscular (IM) dose of mRNA-1273 in SARS-CoV-2 naïve persons. We conducted an open-label, non-inferiority, randomized controlled trial in the Netherlands between June and December 2021. One hundred and fifty healthy and SARS-CoV-2 naïve participants, aged 18-30 years, were randomized (1:1:1) to receive either two doses of 20 µg mRNA-1273 ID with a standard needle (SN) or the Bella-mu® needle (BM), or two doses of 100 µg IM, 28 days apart. The primary outcome was non-inferiority in seroconversion rates at day 43 (D43), defined as a neutralizing antibody concentration threshold of 465 IU/mL, the lowest response in the IM group. The non-inferiority margin was set at -15%. Neutralizing antibody concentrations at D43 were 1789 (95% CI: 1488-2150) in the IM and 1263 (951-1676) and 1295 (1020-1645) in the ID-SN and ID-BM groups, respectively. The absolute difference in seroconversion proportion between fractional and standard-dose groups was -13.95% (-24.31 to -3.60) for the ID-SN and -13.04% (-22.78 to -3.31) for the ID-BM group and exceeded the predefined non-inferiority margin. Although ID vaccination with 1/5th dose of mRNA-1273 did not meet the predefined non-inferior criteria, the neutralizing antibody concentrations in these groups are far above the proposed proxy for protection against severe disease (100 IU/mL), justifying this strategy in times of vaccine scarcity to accelerate mass protection against severe disease.
Collapse
Affiliation(s)
- Manon L M Prins
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Geert V T Roozen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cilia R Pothast
- Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wesley Huisman
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob van Binnendijk
- Department of Immune Surveillance, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Gerco den Hartog
- Department of Immune Surveillance, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Laboratory of Medical Immunology, RadboudUMC, Nijmegen, The Netherlands
| | - Vincent P Kuiper
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Corine Prins
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacqueline J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Olivia A C Lamers
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Pieter R Koopman
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Annelieke C Kruithof
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Center for Human Drug Research, Leiden, The Netherlands
| | - Ingrid M C Kamerling
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Center for Human Drug Research, Leiden, The Netherlands
| | - Romy C Dijkland
- Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alicia C de Kroon
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Shohreh Azimi
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariet C W Feltkamp
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjan Kuijer
- Department of Immune Surveillance, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Simon P Jochems
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mirjam H M Heemskerk
- Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anna H E Roukens
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
40
|
Binayke A, Zaheer A, Vishwakarma S, Singh S, Sharma P, Chandwaskar R, Gosain M, Raghavan S, Murugesan DR, Kshetrapal P, Thiruvengadam R, Bhatnagar S, Pandey AK, Garg PK, Awasthi A. A quest for universal anti-SARS-CoV-2 T cell assay: systematic review, meta-analysis, and experimental validation. NPJ Vaccines 2024; 9:3. [PMID: 38167915 PMCID: PMC10762233 DOI: 10.1038/s41541-023-00794-9] [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: 07/02/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Measuring SARS-CoV-2-specific T cell responses is crucial to understanding an individual's immunity to COVID-19. However, high inter- and intra-assay variability make it difficult to define T cells as a correlate of protection against COVID-19. To address this, we performed systematic review and meta-analysis of 495 datasets from 94 original articles evaluating SARS-CoV-2-specific T cell responses using three assays - Activation Induced Marker (AIM), Intracellular Cytokine Staining (ICS), and Enzyme-Linked Immunospot (ELISPOT), and defined each assay's quantitative range. We validated these ranges using samples from 193 SARS-CoV-2-exposed individuals. Although IFNγ ELISPOT was the preferred assay, our experimental validation suggested that it under-represented the SARS-CoV-2-specific T cell repertoire. Our data indicate that a combination of AIM and ICS or FluoroSpot assay would better represent the frequency, polyfunctionality, and compartmentalization of the antigen-specific T cell responses. Taken together, our results contribute to defining the ranges of antigen-specific T cell assays and propose a choice of assay that can be employed to better understand the cellular immune response against viral diseases.
Collapse
Affiliation(s)
- Akshay Binayke
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
- Jawaharlal Nehru University, New Delhi, India
| | - Aymaan Zaheer
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Siddhesh Vishwakarma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Savita Singh
- Translational Health Science and Technology Institute, Faridabad, India
| | - Priyanka Sharma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Rucha Chandwaskar
- Department of Microbiology, AMITY University Rajasthan, Jaipur, India
| | - Mudita Gosain
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | | | - Ramachandran Thiruvengadam
- Translational Health Science and Technology Institute, Faridabad, India
- Pondicherry Institute of Medical Sciences, Puducherry, India
| | | | | | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, India
- All India Institute of Medical Sciences, New Delhi, India
| | - Amit Awasthi
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India.
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India.
| |
Collapse
|
41
|
Rasche MM, Kaufmann EC, Ratishvili T, Swanson IM, Ovsyannikova IG, Kennedy RB. Detection of SARS-CoV-2-Specific Cells Utilizing Whole Proteins and/or Peptides in Human PBMCs Using IFN-ƴ ELISPOT Assay. Methods Mol Biol 2024; 2768:117-133. [PMID: 38502391 DOI: 10.1007/978-1-0716-3690-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
SARS-CoV-2 continues to threaten global public health, making COVID-19 immunity studies of utmost importance. Waning of antibody responses postinfection and/or vaccination and the emergence of immune escape variants have been ongoing challenges in mitigating SARS-CoV-2 morbidity and mortality. While a tremendous amount of work has been done to characterize humoral immune responses to SARS-CoV-2 virus and vaccines, cellular immunity, mediated by T cells, is critical for efficient viral control and protection and demonstrates high durability and cross-reactivity to coronavirus variants. Thus, ELISPOT, a standard assay for antigen-specific cellular immune response assessment, allows us to evaluate SARS-CoV-2-specific T-cell response by quantifying the frequency of SARS-CoV-2-specific cytokine-secreting cells in vitro. We have outlined a detailed procedure to study T-cell recall responses to SARS-CoV-2 in human peripheral blood mononuclear cells (PBMCs) following infection and/or vaccination using an optimized IFN-γ ELISPOT assay. Our methodologies can be adapted to assess other cytokines and are a useful tool for studying other viral pathogen and/or peptide-specific T-cell responses.
Collapse
Affiliation(s)
| | - Ella C Kaufmann
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Tamar Ratishvili
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | - Ilya M Swanson
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA
| | | | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
42
|
Diego JGB, Singh G, Jangra S, Handrejk K, Laporte M, Chang LA, El Zahed SS, Pache L, Chang MW, Warang P, Aslam S, Mena I, Webb BT, Benner C, García-Sastre A, Schotsaert M. Breakthrough infections by SARS-CoV-2 variants boost cross-reactive hybrid immune responses in mRNA-vaccinated Golden Syrian hamsters. PLoS Pathog 2024; 20:e1011805. [PMID: 38198521 PMCID: PMC10805310 DOI: 10.1371/journal.ppat.1011805] [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: 06/03/2023] [Revised: 01/23/2024] [Accepted: 11/06/2023] [Indexed: 01/12/2024] Open
Abstract
Hybrid immunity (vaccination + natural infection) to SARS-CoV-2 provides superior protection to re-infection. We performed immune profiling studies during breakthrough infections in mRNA-vaccinated hamsters to evaluate hybrid immunity induction. The mRNA vaccine, BNT162b2, was dosed to induce binding antibody titers against ancestral spike, but inefficient serum virus neutralization of ancestral SARS-CoV-2 or variants of concern (VoCs). Vaccination reduced morbidity and controlled lung virus titers for ancestral virus and Alpha but allowed breakthrough infections in Beta, Delta and Mu-challenged hamsters. Vaccination primed for T cell responses that were boosted by infection. Infection back-boosted neutralizing antibody responses against ancestral virus and VoCs. Hybrid immunity resulted in more cross-reactive sera, reflected by smaller antigenic cartography distances. Transcriptomics post-infection reflects both vaccination status and disease course and suggests a role for interstitial macrophages in vaccine-mediated protection. Therefore, protection by vaccination, even in the absence of high titers of neutralizing antibodies in the serum, correlates with recall of broadly reactive B- and T-cell responses.
Collapse
Affiliation(s)
- Juan García-Bernalt Diego
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kim Handrejk
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Manon Laporte
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sara S. El Zahed
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lars Pache
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Max W. Chang
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sadaf Aslam
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Brett T. Webb
- Department of Veterinary Sciences, University of Wyoming, Laramie, Wyoming, United States of America
| | - Christopher Benner
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
43
|
Wang Y, Chen D, Pan Y, Li H, Zhao W, Lu T, Kong W, Ding M, Wang X, Zhang G. Serological response and immune-related adverse events following COVID-19 vaccination in cancer patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Rev Med Virol 2024; 34:e2495. [PMID: 38017632 DOI: 10.1002/rmv.2495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/21/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023]
Abstract
With the popularity of Coronavirus disease 2019 (COVID-19) vaccine and the development of vaccination strategies, the impact of COVID-19 vaccine on cancer patients receiving immune checkpoint inhibitors (ICIs) is still unclear. In the systematic review and meta-analysis of patients with ICIs, we assessed the serological response of cancer patients receiving COVID-19 vaccine, and explored the risk of immune related adverse events (irAEs). We searched PubMed, EMBASE and Cochrane Library as of 10 June 2023, and included cancer patients who received ICIs and COVID-19 vaccine. The systematic review and meta-analysis include cohort study, cross-sectional study and case report. The outcome included the serological response, Spike-specific T-cell response, irAEs and rare adverse events. When possible, the data were analysed by random effect analysis, and the statistical heterogeneity was assessed by Q-test and I2 statistics. We explored the sources of heterogeneity through L'Abbe plots, Galbraith radial plots, and sensitivity analysis. The publication bias was evaluated by Egger's, Begg's linear regression test and funnel plot, and the impact of publication bias was further analysed by trim and fill method. 27 studies were eligible (19 cohort studies, 1 cross-sectional study and 7 case reports), involving 8331 patients (with 4724 receiving ICIs). Most studies used mRNA vaccine (BNT162b2 or mRNA-1273). Compared with cancer patients receiving chemotherapy, cancer patients receiving ICIs were significantly more likely to have seroconversion (RR = 1.05, 95%CI 1.01-1.10, P = 0.02). There were no statistically significant differences in seroconversion rates when comparing cancer patients receiving ICIs with controls without cancer (RR = 0.95, 95% CI 0.89-1.01, P = 0.09) or with cancer patients receiving targeted therapy (RR = 1.05, 95% CI 0.79-1.39, P = 0.75). The incidence of irAEs in patients receiving ICIs before and after COVID-19 vaccination was (21.96%, 95%CI 16.66%-28.94%) and (14.88%, 95%CI 8.65%-25.57%), respectively. The most common irAEs were endocrine abnormalities, skin disorders, etc. The certainty of evidence was low in cancer patients with ICIs, compared with those receiving chemotherapy, and very low versus controls without cancer. Cancer patients treated with ICIs seem to be able to receive COVID-19 vaccine safely without increasing the incidence of irAEs.
Collapse
Affiliation(s)
- Yue Wang
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Dong Chen
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuancan Pan
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Haiming Li
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Weizhe Zhao
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
| | - Taicheng Lu
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Weijia Kong
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Min Ding
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaomin Wang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ganlin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Benoit JM, Breznik JA, Ang JC, Bhakta H, Huynh A, Cowbrough B, Baker B, Heessels L, Lodhi S, Yan E, Ewusie J, Nazy I, Bramson J, Miller MS, Bernatsky S, Larché MJ, Bowdish DME. Immunomodulatory drugs have divergent effects on humoral and cellular immune responses to SARS-CoV-2 vaccination in people living with rheumatoid arthritis. Sci Rep 2023; 13:22846. [PMID: 38129594 PMCID: PMC10739702 DOI: 10.1038/s41598-023-50263-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Understanding the efficacy of SARS-CoV-2 vaccination in people on immunosuppressive drugs, including those with rheumatoid arthritis (RA), is critical for their protection. Vaccine induced protection requires antibodies, CD4+ T cells, and CD8+ T cells, but it is unclear if these are equally affected by immunomodulatory drugs. Here, we determined how humoral and cellular SARS-CoV-2 vaccination responses differed between people with RA and controls, and which drug classes impacted these responses. Blood was collected from participants with RA on immunomodulatory drugs and controls after their second, third, and fourth SARS-CoV-2 vaccinations. Receptor binding domain (RBD)-specific antibodies were quantified by ELISA. Spike-specific memory T cells were quantitated using flow cytometry. Linear mixed models assessed the impact of age, sex, and immunomodulatory drug classes on SARS-CoV-2 vaccination responses. Compared to non-RA controls (n = 35), participants with RA on immunomodulatory drugs (n = 62) had lower anti-RBD IgG and spike-specific CD4+ T cell levels, but no deficits in spike-specific CD8+ T cells, following SARS-CoV-2 vaccination. Use of costimulation inhibitors was associated with lower humoral responses. JAK inhibitors were associated with fewer spike-specific CD4+ T cells. Participants with RA on immunomodulatory drugs mounted weaker responses to SARS-CoV-2 vaccination, with different drug classes impacting the cellular and humoral compartments.
Collapse
Affiliation(s)
- Jenna M Benoit
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Jessica A Breznik
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, ON, Canada
| | - Jann C Ang
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Hina Bhakta
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Angela Huynh
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Braeden Cowbrough
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Barbara Baker
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Lauren Heessels
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sumiya Lodhi
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Elizabeth Yan
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Joycelyne Ewusie
- Department of Health Research Methods, Evidence, and Impact (HEI), McMaster University, Hamilton, ON, Canada
| | - Ishac Nazy
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan Bramson
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Hamilton, ON, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, Hamilton, ON, Canada
- Department of Biochemistry, McMaster University, Hamilton, ON, Canada
| | - Sasha Bernatsky
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Maggie J Larché
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
| | - Dawn M E Bowdish
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- McMaster Immunology Research Centre, Hamilton, ON, Canada.
- Firestone Institute of Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada.
| |
Collapse
|
45
|
Verheul MK, Vos M, de Rond L, De Zeeuw-Brouwer ML, Nijhof KH, Smit D, Oomen D, Molenaar P, Bogaard M, van Bergen R, Middelhof I, Beckers L, Wijmenga-Monsuur AJ, Buisman AM, Boer MC, van Binnendijk R, de Wit J, Guichelaar T. Contribution of SARS-CoV-2 infection preceding COVID-19 mRNA vaccination to generation of cellular and humoral immune responses in children. Front Immunol 2023; 14:1327875. [PMID: 38193077 PMCID: PMC10773747 DOI: 10.3389/fimmu.2023.1327875] [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: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Primary COVID-19 vaccination for children, 5-17 years of age, was offered in the Netherlands at a time when a substantial part of this population had already experienced a SARS-CoV-2 infection. While vaccination has been shown effective, underlying immune responses have not been extensively studied. We studied immune responsiveness to one and/or two doses of primary BNT162b2 mRNA vaccination and compared the humoral and cellular immune response in children with and without a preceding infection. Antibodies targeting the original SARS-CoV-2 Spike or Omicron Spike were measured by multiplex immunoassay. B-cell and T-cell responses were investigated using enzyme-linked immunosorbent spot (ELISpot) assays. The activation of CD4+ and CD8+ T cells was studied by flowcytometry. Primary vaccination induced both a humoral and cellular adaptive response in naive children. These responses were stronger in those with a history of infection prior to vaccination. A second vaccine dose did not further boost antibody levels in those who previously experienced an infection. Infection-induced responsiveness prior to vaccination was mainly detected in CD8+ T cells, while vaccine-induced T-cell responses were mostly by CD4+ T cells. Thus, SARS-CoV-2 infection prior to vaccination enhances adaptive cellular and humoral immune responses to primary COVID-19 vaccination in children. As most children are now expected to contract infection before the age of five, the impact of infection-induced immunity in children is of high relevance. Therefore, considering natural infection as a priming immunogen that enhances subsequent vaccine-responsiveness may help decision-making on the number and timing of vaccine doses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Teun Guichelaar
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
46
|
Masiá M, de la Rica A, Fernández-González M, García JA, Padilla S, García-Abellán J, Botella Á, Mascarell P, Gutiérrez F. Integrating SARS-CoV-2-specific interferon-γ release assay testing in the evaluation of patients hospitalized with COVID-19. Microbiol Spectr 2023; 11:e0241923. [PMID: 37855635 PMCID: PMC10715100 DOI: 10.1128/spectrum.02419-23] [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: 06/08/2023] [Accepted: 09/06/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE The cellular immune response is essential in the protection against severe disease in patients with established SARS-CoV-2 infection. The novelty of this study lies in the evaluation of the overall performance of a standardized assay to measure cellular immune response, the SARS-CoV-2-specific interferon-γ release assay (IGRA), in hospitalized patients with severe COVID-19. The SARS-CoV-2 IGRA was shown to accurately classify patients based on disease severity and prognosis, and the study revealed that test performance was not affected by the SARS-CoV-2 variant or control tube results. We identified an assay cut-off point with a high negative predictive value against mortality. The SARS-CoV-2 IGRA in patients hospitalized for COVID-19 may be a useful tool to assess cellular immunity and adopt targeted therapeutic and preventive measures.
Collapse
Affiliation(s)
- Mar Masiá
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Alba de la Rica
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- Microbiology Service, Hospital General Universitario de Elche, Alicant, Spain
| | - Marta Fernández-González
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - José Alberto García
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Padilla
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Javier García-Abellán
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Ángela Botella
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
| | - Paula Mascarell
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
| | - Félix Gutiérrez
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicant, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Department, Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
47
|
Pérez P, Albericio G, Astorgano D, Flores S, Sánchez-Corzo C, Sánchez-Cordón PJ, Luczkowiak J, Delgado R, Casasnovas JM, Esteban M, García-Arriaza J. Preclinical immune efficacy against SARS-CoV-2 beta B.1.351 variant by MVA-based vaccine candidates. Front Immunol 2023; 14:1264323. [PMID: 38155964 PMCID: PMC10754519 DOI: 10.3389/fimmu.2023.1264323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
The constant appearance of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VoCs) has jeopardized the protective capacity of approved vaccines against coronavirus disease-19 (COVID-19). For this reason, the generation of new vaccine candidates adapted to the emerging VoCs is of special importance. Here, we developed an optimized COVID-19 vaccine candidate using the modified vaccinia virus Ankara (MVA) vector to express a full-length prefusion-stabilized SARS-CoV-2 spike (S) protein, containing 3 proline (3P) substitutions in the S protein derived from the beta (B.1.351) variant, termed MVA-S(3Pbeta). Preclinical evaluation of MVA-S(3Pbeta) in head-to-head comparison to the previously generated MVA-S(3P) vaccine candidate, expressing a full-length prefusion-stabilized Wuhan S protein (with also 3P substitutions), demonstrated that two intramuscular doses of both vaccine candidates fully protected transgenic K18-hACE2 mice from a lethal challenge with SARS-CoV-2 beta variant, reducing mRNA and infectious viral loads in the lungs and in bronchoalveolar lavages, decreasing lung histopathological lesions and levels of proinflammatory cytokines in the lungs. Vaccination also elicited high titers of anti-S Th1-biased IgGs and neutralizing antibodies against ancestral SARS-CoV-2 Wuhan strain and VoCs alpha, beta, gamma, delta, and omicron. In addition, similar systemic and local SARS-CoV-2 S-specific CD4+ and CD8+ T-cell immune responses were elicited by both vaccine candidates after a single intranasal immunization in C57BL/6 mice. These preclinical data support clinical evaluation of MVA-S(3Pbeta) and MVA-S(3P), to explore whether they can diversify and potentially increase recognition and protection of SARS-CoV-2 VoCs.
Collapse
Affiliation(s)
- Patricia Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Guillermo Albericio
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - David Astorgano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Sara Flores
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Cristina Sánchez-Corzo
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pedro J. Sánchez-Cordón
- Pathology Department, Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Joanna Luczkowiak
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Delgado
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - José M. Casasnovas
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
48
|
Mohazzab A, Fallah Mehrabadi MH, Es-Haghi A, Kalantari S, Mokhberalsafa L, Setarehdan SA, Sadeghi F, Rezaei Mokarram A, Haji Moradi M, Razaz SH, Taghdiri M, Ansarifar A, Lotfi M, Khorasani A, Nofeli M, Masoumi S, Boluki Z, Erfanpoor S, Bagheri Amiri F, Esmailzadehha N, Filsoof S, Mohseni V, Ghahremanzadeh N, Safari S, Shahsavan M, Bayazidi S, Raghami Derakhshani M, Rabiee MH, Golmoradi-Zadeh R, Khodadoost B, Solaymani-Dodaran M, Banihashemi SR. Phase II, Safety and Immunogenicity of RAZI Cov Pars (RCP) SARS Cov-2 Vaccine in Adults Aged 18-70 Years; A Randomized, Double-Blind Clinical Trial. J Pharm Sci 2023; 112:3012-3021. [PMID: 37832918 DOI: 10.1016/j.xphs.2023.09.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/30/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND This study explores the safety and immunogenicity of the Razi-Cov-Pars (RCP) SARS Cov-2 recombinant spike protein vaccine. METHOD In a randomized, double-blind, placebo-controlled trial, adults aged 18-70 were randomly allocated to receive selected 10 µg/200 µl vaccine strengths or placebo (adjuvant). It included two intramuscular injections at days 0 and 21, followed by an intranasal dose at day 51. Immediate and delayed solicited local and systemic adverse reactions after each dose up to a week, and specific IgG antibodies against SARS Cov-2 spike antigens two weeks after the 2nd dose were assessed as primary outcomes. Secondary safety outcomes were abnormal laboratory findings and medically attended adverse events (MAAE) over six months follow up. Secondary immunogenicity outcomes were neutralizing antibody activity and cell-mediated immune response. RESULT Between May 27th and July 15th, 2021, 500 participants were enrolled. Participants' mean (SD) age was 37.8 (9.0), and 67.0 % were male. No immediate adverse reaction was observed following the intervention. All solicited local and systemic adverse events were moderate (Grade I-II). Specific IgG antibody response against S antigen in the vaccine group was 5.28 times (95 %CI: 4.02-6.94) the placebo group with a 75 % seroconversion rate. During six months of follow-up, 8 SAEs were reported, unrelated to the study intervention. The participants sustained their acquired humoral responses at the end of the sixth month. The vaccine predominantly resulted in T-helper 1 cell-mediated immunity, CD8+ cytotoxic T-cell increase, and no increase in inflammatory IL-6 cytokine. CONCLUSION RCP vaccine is safe and creates strong and durable humoral and cellular immunity. TRIAL REGISTRATION (IRCT20201214049709N2).
Collapse
Affiliation(s)
- Arash Mohazzab
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Reproductive Biotechnology Research Center, Avicenna Research Institute Tehran, ACECR, Tehran, Iran
| | - Mohammad Hossein Fallah Mehrabadi
- Department of epidemiology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Ali Es-Haghi
- Department of Physico Chemistry, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Saeed Kalantari
- Departments of Infectious Diseases and Tropical Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ladan Mokhberalsafa
- Department of epidemiology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | | | - Fariba Sadeghi
- Department of epidemiology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Ali Rezaei Mokarram
- Research and Development Department, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Monireh Haji Moradi
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Seyad Hossein Razaz
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Maryam Taghdiri
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Akram Ansarifar
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Lotfi
- Department of Quality Control, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Akbar Khorasani
- Research and Development Department, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mojtaba Nofeli
- Research and Development Department, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Safdar Masoumi
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Boluki
- Knowledge Utilization Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Erfanpoor
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Bagheri Amiri
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging infectious diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Neda Esmailzadehha
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Filsoof
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Vahideh Mohseni
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Shiva Safari
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Shahsavan
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Shnoo Bayazidi
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Raghami Derakhshani
- Department of epidemiology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Mohammad Hasan Rabiee
- Department Of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Rezvan Golmoradi-Zadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnam Khodadoost
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Solaymani-Dodaran
- Clinical Trial Center, Iran University of Medical Science, Tehran, Iran; Minimally Invasive Surgery Research Center, Hazrat-e-Rasool Hospital, Iran University of Medical Science, Tehran, Iran.
| | - Seyed Reza Banihashemi
- Department of immunology, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran.
| |
Collapse
|
49
|
Fedele G, Schiavoni I, Trentini F, Leone P, Olivetta E, Fallucca A, Fiore S, Di Martino A, Abrignani S, Baldo V, Baldovin T, Bandera A, Clerici P, De Paschale M, Diaco F, Domnich A, Fortunato F, Giberti I, Gori A, Grifantini R, Lazzarotto T, Lodi V, Mastroianni CM, Prato R, Restivo V, Vitale F, Brusaferro S, Merler S, Palamara AT, Stefanelli P. A 12-month follow-up of the immune response to SARS-CoV-2 primary vaccination: evidence from a real-world study. Front Immunol 2023; 14:1272119. [PMID: 38077369 PMCID: PMC10698351 DOI: 10.3389/fimmu.2023.1272119] [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: 08/03/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
A real-world population-based longitudinal study, aimed at determining the magnitude and duration of immunity induced by different types of vaccines against COVID-19, started in 2021 by enrolling a cohort of 2,497 individuals at time of their first vaccination. The study cohort included both healthy adults aged ≤65 years and elderly subjects aged >65 years with two or more co-morbidities. Here, patterns of anti-SARS-CoV-2 humoral and cell-mediated specific immune response, assessed on 1,182 remaining subjects, at 6 (T6) and 12 months (T12) after the first vaccine dose, are described. At T12 median anti-Spike IgG antibody levels were increased compared to T6. The determinants of increased anti-Spike IgG were the receipt of a third vaccine dose between T6 and T12 and being positive for anti-Nucleocapside IgG at T12, a marker of recent infection, while age had no significant effect. The capacity of T12 sera to neutralize in vitro the ancestral B strain and the Omicron BA.5 variant was assessed in a subgroup of vaccinated subjects. A correlation between anti-S IgG levels and sera neutralizing capacity was identified and higher neutralizing capacity was evident in healthy adults compared to frail elderly subjects and in those who were positive for anti-Nucleocapside IgG at T12. Remarkably, one third of T12 sera from anti-Nucleocapside IgG negative older individuals were unable to neutralize the BA.5 variant strain. Finally, the evaluation of T-cell mediated immunity showed that most analysed subjects, independently from age and comorbidity, displayed Spike-specific responses with a high degree of polyfunctionality, especially in the CD8 compartment. In conclusion, vaccinated subjects had high levels of circulating antibodies against SARS-CoV-2 Spike protein 12 months after the primary vaccination, which increased as compared to T6. The enhancing effect could be attributable to the administration of a third vaccine dose but also to the occurrence of breakthrough infection. Older individuals, especially those who were anti-Nucleocapside IgG negative, displayed an impaired capacity to neutralize the BA.5 variant strain. Spike specific T-cell responses, able to sustain immunity and maintain the ability to fight the infection, were present in most of older and younger subjects assayed at T12.
Collapse
Affiliation(s)
- Giorgio Fedele
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Ilaria Schiavoni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Filippo Trentini
- Center for Health Emergencies, Bruno Kessler Foundation, Trento, Italy
- Dondena Centre for Research on Social Dynamics and Public Policy, Bocconi University, Milan, Italy
| | - Pasqualina Leone
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Olivetta
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Fallucca
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Stefano Fiore
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Angela Di Martino
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Sergio Abrignani
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
- Department of Clinical Sciences & Community Health, University of Milan, Milan, Italy
| | - Vincenzo Baldo
- Laboratory of Hygiene and Applied Microbiology, Hygiene and Public Health Unit, Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Tatjana Baldovin
- Laboratory of Hygiene and Applied Microbiology, Hygiene and Public Health Unit, Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milan, Italy
| | - Pierangelo Clerici
- Microbiology Unit, Azienda Socio Sanitaria Territoriale (ASST) Ovest Milanese, Milan, Italy
| | - Massimo De Paschale
- Microbiology Unit, Azienda Socio Sanitaria Territoriale (ASST) Ovest Milanese, Milan, Italy
| | - Fabiana Diaco
- Department of Molecular Medicine, AOU Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Alexander Domnich
- IRCCS Ospedale Policlinico San Martino Genova, and Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Francesca Fortunato
- Hygiene Unit, Policlinico Riuniti Foggia Hospital, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Irene Giberti
- IRCCS Ospedale Policlinico San Martino Genova, and Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Andrea Gori
- Microbiology Unit, Azienda Socio Sanitaria Territoriale (ASST) Ovest Milanese, Milan, Italy
- II Division of Infectious Diseases, "Luigi Sacco" Hospital, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
| | - Tiziana Lazzarotto
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Section of Microbiology, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vittorio Lodi
- Occupational Health Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Disease, AOU Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Rosa Prato
- IRCCS Ospedale Policlinico San Martino Genova, and Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Vincenzo Restivo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Francesco Vitale
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | | | - Stefano Merler
- Center for Health Emergencies, Bruno Kessler Foundation, Trento, Italy
| | | | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
50
|
Moreno LB, de Albuquerque JB, Neary JM, Nguyen T, Hastie KM, Landeras-Bueno S, Hariharan C, Nathan A, Getz MA, Gayton AC, Khatri A, Gaiha GD, Saphire EO, Luster AD, Moon JJ. Identification of mouse CD4 + T cell epitopes in SARS-CoV-2 BA.1 spike and nucleocapsid for use in peptide:MHCII tetramers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.566918. [PMID: 38014059 PMCID: PMC10680761 DOI: 10.1101/2023.11.16.566918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Understanding adaptive immunity against SARS-CoV-2 is a major requisite for the development of effective vaccines and treatments for COVID-19. CD4+ T cells play an integral role in this process primarily by generating antiviral cytokines and providing help to antibody-producing B cells. To empower detailed studies of SARS-CoV-2-specific CD4+ T cell responses in mouse models, we comprehensively mapped I-Ab-restricted epitopes for the spike and nucleocapsid proteins of the BA.1 variant of concern via IFNγ ELISpot assay. This was followed by the generation of corresponding peptide:MHCII tetramer reagents to directly stain epitope-specific T cells. Using this rigorous validation strategy, we identified 6 reliably immunogenic epitopes in spike and 3 in nucleocapsid, all of which are conserved in the ancestral Wuhan strain. We also validated a previously identified epitope from Wuhan that is absent in BA.1. These epitopes and tetramers will be invaluable tools for SARS-CoV-2 antigen-specific CD4+ T cell studies in mice.
Collapse
Affiliation(s)
- Laura Bricio Moreno
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Juliana Barreto de Albuquerque
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Jake M. Neary
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
| | - Thao Nguyen
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
| | - Kathryn M. Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Sara Landeras-Bueno
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Chitra Hariharan
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Anusha Nathan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA, United States
| | - Matthew A. Getz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Alton C. Gayton
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Ashok Khatri
- Harvard Medical School, Boston, MA, United States
- Endocrine Division, Massachusetts General Hospital, Boston, MA, United States
| | - Gaurav D. Gaiha
- Harvard Medical School, Boston, MA, United States
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, United States
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Andrew D. Luster
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - James J. Moon
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, United States
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|