1
|
Grant M, Ni Lee L, Chinnakannan S, Tong O, Kwok J, Cianci N, Tillman L, Saha A, Pereira Almeida V, Leung C. Unlocking cancer vaccine potential: What are the key factors? Hum Vaccin Immunother 2024; 20:2331486. [PMID: 38564321 DOI: 10.1080/21645515.2024.2331486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer is a global health challenge, with changing demographics and lifestyle factors producing an increasing burden worldwide. Screening advancements are enabling earlier diagnoses, but current cancer immunotherapies only induce remission in a small proportion of patients and come at a high cost. Cancer vaccines may offer a solution to these challenges, but they have been mired by poor results in past decades. Greater understanding of tumor biology, coupled with the success of vaccine technologies during the COVID-19 pandemic, has reinvigorated cancer vaccine development. With the first signs of efficacy being reported, cancer vaccines may be beginning to fulfill their potential. Solid tumors, however, present different hurdles than infectious diseases. Combining insights from previous cancer vaccine clinical development and contemporary knowledge of tumor immunology, we ask: who are the 'right' patients, what are the 'right' targets, and which are the 'right' modalities to maximize the chances of cancer vaccine success?
Collapse
|
2
|
Sheetikov SA, Khmelevskaya AA, Zornikova KV, Zvyagin IV, Shomuradova AS, Serdyuk YV, Shakirova NT, Peshkova IO, Titov A, Romaniuk DS, Shagina IA, Chudakov DM, Kiryukhin DO, Shcherbakova OV, Khamaganova EG, Dzutseva V, Afanasiev A, Bogolyubova AV, Efimov GA. Clonal structure and the specificity of vaccine-induced T cell response to SARS-CoV-2 Spike protein. Front Immunol 2024; 15:1369436. [PMID: 38629062 PMCID: PMC11018901 DOI: 10.3389/fimmu.2024.1369436] [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: 01/12/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Adenovirus vaccines, particularly the COVID-19 Ad5-nCoV adenovirus vaccine, have emerged as promising tools in the fight against infectious diseases. In this study, we investigated the structure of the T cell response to the Spike protein of the SARS-CoV-2 virus used in the COVID-19 Ad5-nCoV adenoviral vaccine in a phase 3 clinical trial (NCT04540419). In 69 participants, we collected peripheral blood samples at four time points after vaccination or placebo injection. Sequencing of T cell receptor repertoires from Spike-stimulated T cell cultures at day 14 from 17 vaccinated revealed a more diverse CD4+ T cell repertoire compared to CD8+. Nevertheless, CD8+ clonotypes accounted for more than half of the Spike-specific repertoire. Our longitudinal analysis showed a peak T cell response at day 14, followed by a decline until month 6. Remarkably, multiple T cell clonotypes persisted for at least 6 months after vaccination, as demonstrated by ex vivo stimulation. Examination of CDR3 regions revealed homologous sequences in both CD4+ and CD8+ clonotypes, with major CD8+ clonotypes sharing high similarity with annotated sequences specific for the NYNYLYRLF peptide, suggesting potential immunodominance. In conclusion, our study demonstrates the immunogenicity of the Ad5-nCoV adenoviral vaccine and highlights its ability to induce robust and durable T cell responses. These findings provide valuable insight into the efficacy of the vaccine against COVID-19 and provide critical information for ongoing efforts to control infectious diseases.
Collapse
Affiliation(s)
- Saveliy A. Sheetikov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra A. Khmelevskaya
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Ksenia V. Zornikova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Ivan V. Zvyagin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Alina S. Shomuradova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana V. Serdyuk
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Naina T. Shakirova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Iuliia O. Peshkova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Aleksei Titov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Dmitrii S. Romaniuk
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Irina A. Shagina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Dmitry M. Chudakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Dmitry O. Kiryukhin
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Olga V. Shcherbakova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Ekaterina G. Khamaganova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Vitalina Dzutseva
- Novosibirsk State University, Medical School, Novosibirsk, Russia
- NPO Petrovax Pharm LLC, Moscow, Russia
| | | | | | - Grigory A. Efimov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| |
Collapse
|
3
|
Vogrig A, Tartaglia S, Dentoni M, Fabris M, Bax F, Belluzzo M, Verriello L, Bagatto D, Gastaldi M, Tocco P, Zoccarato M, Zuliani L, Pilotto A, Padovani A, Villagrán-García M, Davy V, Gigli GL, Honnorat J, Valente M. Central nervous system immune-related disorders after SARS-CoV-2 vaccination: a multicenter study. Front Immunol 2024; 15:1344184. [PMID: 38375477 PMCID: PMC10876052 DOI: 10.3389/fimmu.2024.1344184] [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: 11/25/2023] [Accepted: 01/09/2024] [Indexed: 02/21/2024] Open
Abstract
Background COVID-19 vaccines have been approved due to their excellent safety and efficacy data and their use has also permitted to reduce neurological complications of SARS-CoV-2. However, clinical trials were underpowered to detect rare adverse events. Herein, the aim was to characterize the clinical spectrum and immunological features of central nervous system (CNS) immune-related events following SARS-CoV-2 vaccination. Methods Multicenter, retrospective, cohort study (December 1, 2020-April 30, 2022). Inclusion criteria were (1) de novo CNS disorders developing after SARS-CoV-2 vaccination (probable causal relationship as per 2021 Butler criteria) (2); evidence for an immune-mediated etiology, as per (i) 2016 Graus criteria for autoimmune encephalitis (AE); (ii) 2015 Wingerchuk criteria for neuromyelitis optica spectrum disorders; (iii) criteria for myelitis. Results Nineteen patients were included from 7 tertiary referral hospitals across Italy and France (one of them being a national referral center for AE), over almost 1 year and half of vaccination campaign. Vaccines administered were mRNA-based (63%) and adenovirus-vectored (37%). The median time between vaccination and symptoms onset was 14 days (range: 2-41 days). CSF was inflammatory in 74%; autoantibodies were detected in 5%. CSF cytokine analysis (n=3) revealed increased CXCL-10 (IP-10), suggesting robust T-cell activation. The patients had AE (58%), myelitis (21%), acute disseminated encephalomyelitis (ADEM) (16%), and brainstem encephalitis (5%). All patients but 2 received immunomodulatory treatment. At last follow-up (median 130 days; range: 32-540), only one patient (5%) had a mRS>2. Conclusion CNS adverse events of COVID-19 vaccination appear to be very rare even at reference centers and consist mostly of antibody-negative AE, myelitis, and ADEM developing approximately 2 weeks after vaccination. Most patients improve following immunomodulatory treatment.
Collapse
Affiliation(s)
- Alberto Vogrig
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Sara Tartaglia
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Marta Dentoni
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Martina Fabris
- Institute of Clinical Pathology, Department of Laboratory Medicine, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Francesco Bax
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Marco Belluzzo
- Neurology Unit, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Lorenzo Verriello
- Neurology Unit, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Daniele Bagatto
- Department of Diagnostic Imaging, Unit of Neuroradiology, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Matteo Gastaldi
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| | - Pierluigi Tocco
- Neurology and Stroke Unit, “Spirito Santo” Hospital of Pescara, Pescara, Italy
| | - Marco Zoccarato
- UOC Neurologia O.S.A. - Azienda Ospedale Università di Padova, Padua, Italy
| | - Luigi Zuliani
- Neurology Unit, AULSS8 Berica, San Bortolo Hospital, Vicenza, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia University Hospital, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Continuity of Care and Frailty, ASST Spedali Civili Brescia University Hospital, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
| | - Macarena Villagrán-García
- French Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
| | - Vincent Davy
- Department of Neurology, Hôpital Pitié Salpétrière, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Gian Luigi Gigli
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Jérôme Honnorat
- French Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique, Bron, France
- MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Université Claude Bernard Lyon 1, Lyon, France
| | - Mariarosaria Valente
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Clinical Neurology, Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| |
Collapse
|
4
|
Safont G, Villar-Hernández R, Smalchuk D, Stojanovic Z, Marín A, Lacoma A, Pérez-Cano C, López-Martínez A, Molina-Moya B, Solis AJ, Arméstar F, Matllo J, Díaz-Fernández S, Romero I, Casas I, Strecker K, Preyer R, Rosell A, Latorre I, Domínguez J. Measurement of IFN-γ and IL-2 for the assessment of the cellular immunity against SARS-CoV-2. Sci Rep 2024; 14:1137. [PMID: 38212416 PMCID: PMC10784529 DOI: 10.1038/s41598-024-51505-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: 07/17/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024] Open
Abstract
The study of specific T-cell responses against SARS-CoV-2 is important for understanding long-term immunity and infection management. The aim of this study was to assess the dual IFN-γ and IL-2 detection, using a SARS-CoV-2 specific fluorescence ELISPOT, in patients undergoing acute disease, during convalescence, and after vaccination. We also evaluated humoral response and compared with T-cells with the aim of correlating both types of responses, and increase the number of specific response detection. Blood samples were drawn from acute COVID-19 patients and convalescent individuals classified according to disease severity; and from unvaccinated and vaccinated uninfected individuals. IgGs against Spike and nucleocapsid, IgMs against nucleocapsid, and neutralizing antibodies were also analyzed. Our results show that IFN-γ in combination with IL-2 increases response detection in acute and convalescent individuals (p = 0.023). In addition, IFN-γ detection can be a useful biomarker for monitoring severe acute patients, as our results indicate that those individuals with a poor outcome have lower levels of this cytokine. In some cases, the lack of cellular immunity is compensated by antibodies, confirming the role of both types of immune responses in infection, and confirming that their dual detection can increase the number of specific response detections. In summary, IFN-γ/IL-2 dual detection is promising for characterizing and assessing the immunization status, and helping in the patient management.
Collapse
Affiliation(s)
- Guillem Safont
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Raquel Villar-Hernández
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Daria Smalchuk
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- Odesa I. I. Mechnykov National University, Odesa, Ukraine
| | - Zoran Stojanovic
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alicia Marín
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alicia Lacoma
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Pérez-Cano
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Anabel López-Martínez
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Bárbara Molina-Moya
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alan Jhunior Solis
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Fernando Arméstar
- Intensive Care Medicine Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Joan Matllo
- Basic Unit for the Prevention of Occupational Risks (UBP), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Sergio Díaz-Fernández
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Iris Romero
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irma Casas
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Preventive Medicine Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Kevin Strecker
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Rosemarie Preyer
- Genome Identification Diagnostics GmbH (GenID), Straßberg, Germany
| | - Antoni Rosell
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
- Pulmonology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Irene Latorre
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose Domínguez
- Institut d'Investigació Germans Trias i Pujol, Barcelona, Spain.
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
- Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
5
|
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
|
6
|
Nam M, Cha JH, Kim SW, Kim SB, Lee KB, Chung YS, Yun SG, Nam MH, Lee CK, Cho Y. Performance Evaluation of Three Antibody Binding Assays, a Neutralizing Antibody Assay, and an Interferon-Gamma Release Assay for SARS-CoV-2 According to Vaccine Type in Vaccinated Group. Diagnostics (Basel) 2023; 13:3688. [PMID: 38132272 PMCID: PMC10742828 DOI: 10.3390/diagnostics13243688] [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: 10/10/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
We evaluated the performance of SARS-CoV-2 assays in the vaccinated group using receptor-binding domain antibody assays (RBD Ab assay), neutralizing antibody assay (nAb assay), and interferon-gamma release assay (IGR assay). We also compared the performance of the SARS-CoV-2 assays based on vaccine type in a large population. We collected 1851 samples from vaccinated individuals with vector, mix-and-match (MM), and mRNA vaccines. The performance of the RBD Ab assays was assessed by SARS-CoV-2 IgG II Quant (Abbott Laboratories, Sligo, Ireland), SARS-CoV-2 IgG (Beckman Coulter, CA, USA), and anti-SARS-CoV-2 S (Roche Diagnostics GmbH, Mannheim, Germany). The nAb assay was assessed by cPass SARS-CoV-2 neutralization antibody detection kits (GenScript, NJ, USA). The IGR assay was assessed by QuantiFERON (Qiagen, Venlo, The Netherlands). Median values of the RBD Ab assays and nAb assay sequentially increased after the first and second vaccinations. RBD Ab assays and nAb assay showed very strong correlations. The median values of the RBD Ab, nAb, and IGR were higher in the mRNA vaccine group than in the vector and MM vaccine groups. The agreement and correlation among the RBD Ab assays, nAb assay, and IGR assay were higher in the mRNA vaccine group than in the vector and MM vaccine groups. We compared the performance of the RBD Ab assay, nAb assay, and IGR assay based on the vaccine types using the RBD Ab, nAb, and IGR assays. This study provides a better understanding of the assessment of humoral and cellular immune responses after vaccination.
Collapse
Affiliation(s)
- Minjeong Nam
- Department of Laboratory Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (M.N.); (S.G.Y.); (M.-H.N.); (C.K.L.)
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (J.H.C.); (S.-W.K.)
| | - Jae Hyun Cha
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (J.H.C.); (S.-W.K.)
| | - Sang-Wook Kim
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (J.H.C.); (S.-W.K.)
| | - Sun Bean Kim
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (S.B.K.); (K.-B.L.); (Y.-S.C.)
| | - Ki-Byung Lee
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (S.B.K.); (K.-B.L.); (Y.-S.C.)
| | - You-Seung Chung
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (S.B.K.); (K.-B.L.); (Y.-S.C.)
| | - Seung Gyu Yun
- Department of Laboratory Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (M.N.); (S.G.Y.); (M.-H.N.); (C.K.L.)
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (J.H.C.); (S.-W.K.)
| | - Myung-Hyun Nam
- Department of Laboratory Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (M.N.); (S.G.Y.); (M.-H.N.); (C.K.L.)
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (J.H.C.); (S.-W.K.)
| | - Chang Kyu Lee
- Department of Laboratory Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (M.N.); (S.G.Y.); (M.-H.N.); (C.K.L.)
| | - Yunjung Cho
- Department of Laboratory Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (M.N.); (S.G.Y.); (M.-H.N.); (C.K.L.)
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul 02841, Republic of Korea; (J.H.C.); (S.-W.K.)
| |
Collapse
|
7
|
Ferentinos P, Snape D, Koivula F, Faustini S, Nicholson-Little A, Stacey M, Gifford R, Parsons I, Lamb L, Greeves J, O'Hara J, Cunningham AF, Woods D, Richter A, O'Shea MK. Validation of dried blood spot sampling for detecting SARS-CoV-2 antibodies and total immunoglobulins in a large cohort of asymptomatic young adults. J Immunol Methods 2023; 518:113492. [PMID: 37201783 DOI: 10.1016/j.jim.2023.113492] [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: 12/19/2022] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Detecting antibody responses following infection with SARS-CoV-2 is necessary for sero-epidemiological studies and assessing the role of specific antibodies in disease, but serum or plasma sampling is not always viable due to logistical challenges. Dried blood spot sampling (DBS) is a cheaper, simpler alternative and samples can be self-collected and returned by post, reducing risk for SARS-CoV-2 exposure from direct patient contact. The value of large-scale DBS sampling for the assessment of serological responses to SARS-CoV-2 has not been assessed in depth and provides a model for examining the logistics of using this approach to other infectious diseases. The ability to measure specific antigens is attractive for remote outbreak situations where testing may be limited or for patients who require sampling after remote consultation. METHODS We compared the performance of SARS-CoV-2 anti-spike and anti-nucleocapsid antibody detection from DBS samples with matched serum collected by venepuncture in a large population of asymptomatic young adults (N = 1070) living and working in congregate settings (military recruits, N = 625); university students, N = 445). We also compared the effect of self-sampling (ssDBS) with investigator-collected samples (labDBS) on assay performance, and the quantitative measurement of total IgA, IgG and IgM between DBS eluates and serum. RESULTS Baseline seropositivity for anti-Spike IgGAM antibody was significantly higher among university students than military recruits. Strong correlations were observed between matched DBS and serum samples in both university students and recruits for the anti-spike IgGAM assay. Minimal differences were found in results by ssDBS and labDBS and serum by Bland Altman and Cohen kappa analyses. LabDBS achieved 82.0% sensitivity and 98.2% specificity and ssDBS samples 86.1% sensitivity and 96.7% specificity for detecting anti-Spike IgGAM antibodies relative to serum samples. For anti-SARS-CoV-2 nucleocapsid IgG there was qualitatively 100% agreement between serum and DBS samples and weak correlation in ratio measurements. Strong correlations were observed between serum and DBS-derived total IgG, IgA, and IgM. CONCLUSIONS This is the largest validation of DBS against paired serum for SARS-CoV-2 specific antibody measurement and we have shown that DBS retains performance from prior smaller studies. There were no significant differences regarding DBS collection methods, suggesting that self-collected samples are a viable sampling collection method. These data offer confidence that DBS can be employed more widely as an alternative to classical serology.
Collapse
Affiliation(s)
- P Ferentinos
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK
| | - D Snape
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK
| | - F Koivula
- Department of Army Health and Performance Research, Andover, Hampshire, UK
| | - S Faustini
- Clinical Immunology Service, University of Birmingham, Birmingham, UK
| | - A Nicholson-Little
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK
| | - M Stacey
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK; Research & Clinical Innovation, Royal Centre for Defence Medicine, Birmingham, UK
| | - R Gifford
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK; Research & Clinical Innovation, Royal Centre for Defence Medicine, Birmingham, UK
| | - I Parsons
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK; Research & Clinical Innovation, Royal Centre for Defence Medicine, Birmingham, UK
| | - L Lamb
- Research & Clinical Innovation, Royal Centre for Defence Medicine, Birmingham, UK
| | - J Greeves
- Department of Army Health and Performance Research, Andover, Hampshire, UK
| | - J O'Hara
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK
| | - A F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - D Woods
- Research Institute for Sport, Physical Activity and Leisure, Carnegie School of Sport, Leeds Beckett University, UK; Research & Clinical Innovation, Royal Centre for Defence Medicine, Birmingham, UK
| | - A Richter
- Clinical Immunology Service, University of Birmingham, Birmingham, UK; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - M K O'Shea
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; Research & Clinical Innovation, Royal Centre for Defence Medicine, Birmingham, UK.
| |
Collapse
|
8
|
Shahjin F, Patel M, Machhi J, Cohen JD, Nayan MU, Yeapuri P, Zhang C, Waight E, Hasan M, Abdelmoaty MM, Dash PK, Zhou Y, Andreu I, Gendelman HE, Kevadiya BD. Multipolymer microsphere delivery of SARS-CoV-2 antigens. Acta Biomater 2023; 158:493-509. [PMID: 36581007 PMCID: PMC9791794 DOI: 10.1016/j.actbio.2022.12.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/08/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Effective antigen delivery facilitates antiviral vaccine success defined by effective immune protective responses against viral exposures. To improve severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antigen delivery, a controlled biodegradable, stable, biocompatible, and nontoxic polymeric microsphere system was developed for chemically inactivated viral proteins. SARS-CoV-2 proteins encapsulated in polymeric microspheres induced robust antiviral immunity. The viral antigen-loaded microsphere system can preclude the need for repeat administrations, highlighting its potential as an effective vaccine. STATEMENT OF SIGNIFICANCE: Successful SARS-CoV-2 vaccines were developed and quickly approved by the US Food and Drug Administration (FDA). However, each of the vaccines requires boosting as new variants arise. We posit that injectable biodegradable polymers represent a means for the sustained release of emerging viral antigens. The approach offers a means to reduce immunization frequency by predicting viral genomic variability. This strategy could lead to longer-lasting antiviral protective immunity. The current proof-of-concept multipolymer study for SARS-CoV-2 achieve these metrics.
Collapse
Affiliation(s)
- Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Jacob D Cohen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Mohammad Ullah Nayan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Chen Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Emiko Waight
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Prasanta K Dash
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - You Zhou
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Irene Andreu
- RI Consortium of Nanoscience and Nanotechnology and Department of Chemical Engineering University of Rhode Island, RI, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA.
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| |
Collapse
|
9
|
Opposite Effects of mRNA-Based and Adenovirus-Vectored SARS-CoV-2 Vaccines on Regulatory T Cells: A Pilot Study. Biomedicines 2023; 11:biomedicines11020511. [PMID: 36831046 PMCID: PMC9953737 DOI: 10.3390/biomedicines11020511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/12/2023] Open
Abstract
New-generation mRNA and adenovirus vectored vaccines against SARS-CoV-2 spike protein are endowed with immunogenic, inflammatory and immunomodulatory properties. Recently, BioNTech developed a noninflammatory tolerogenic mRNA vaccine (MOGm1Ψ) that induces in mice robust expansion of antigen-specific regulatory T (Treg) cells. The Pfizer/BioNTech BNT162b2 mRNA vaccine against SARS-CoV-2 is identical to MOGm1Ψ except for the lipid carrier, which differs for containing lipid nanoparticles rather than lipoplex. Here we report that vaccination with BNT162b2 led to an increase in the frequency and absolute count of CD4posCD25highCD127low putative Treg cells; in sharp contrast, vaccination with the adenovirus-vectored ChAdOx1 nCoV-19 vaccine led to a significant decrease of CD4posCD25high cells. This pilot study is very preliminary, suffers from important limitations and, frustratingly, very hardly can be refined in Italy because of the >90% vaccination coverage. Thus, the provocative perspective that BNT162b2 and MOGm1Ψ may share the capacity to promote expansion of Treg cells deserves confirmatory studies in other settings.
Collapse
|
10
|
Lo Tartaro D, Paolini A, Mattioli M, Swatler J, Neroni A, Borella R, Santacroce E, Di Nella A, Gozzi L, Busani S, Cuccorese M, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Piwocka K, Gibellini L, Cossarizza A, De Biasi S. Detailed characterization of SARS-CoV-2-specific T and B cells after infection or heterologous vaccination. Front Immunol 2023; 14:1123724. [PMID: 36845156 PMCID: PMC9947839 DOI: 10.3389/fimmu.2023.1123724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
The formation of a robust long-term antigen (Ag)-specific memory, both humoral and cell-mediated, is created following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination. Here, by using polychromatic flow cytometry and complex data analyses, we deeply investigated the magnitude, phenotype, and functionality of SARS-CoV-2-specific immune memory in two groups of healthy subjects after heterologous vaccination compared to a group of subjects who recovered from SARS-CoV-2 infection. We find that coronavirus disease 2019 (COVID-19) recovered patients show different long-term immunological profiles compared to those of donors who had been vaccinated with three doses. Vaccinated individuals display a skewed T helper (Th)1 Ag-specific T cell polarization and a higher percentage of Ag-specific and activated memory B cells expressing immunoglobulin (Ig)G compared to those of patients who recovered from severe COVID-19. Different polyfunctional properties characterize the two groups: recovered individuals show higher percentages of CD4+ T cells producing one or two cytokines simultaneously, while the vaccinated are distinguished by highly polyfunctional populations able to release four molecules, namely, CD107a, interferon (IFN)-γ, tumor necrosis factor (TNF), and interleukin (IL)-2. These data suggest that functional and phenotypic properties of SARS-CoV-2 adaptive immunity differ in recovered COVID-19 individuals and vaccinated ones.
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Julian Swatler
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Alessia Di Nella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Licia Gozzi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Michela Cuccorese
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- National Institute for Cardiovascular Research, Bologna, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| |
Collapse
|
11
|
Shishido AA, Barnes AH, Narayanan S, Chua JV. COVID-19 Vaccines-All You Want to Know. Semin Respir Crit Care Med 2023; 44:143-172. [PMID: 36646092 DOI: 10.1055/s-0042-1759779] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic has led to an unprecedented public health crisis. The collective global response has led to production of multiple safe and effective vaccines utilizing novel platforms to combat the virus that have propelled the field of vaccinology forward. Significant challenges to universal vaccine effectiveness remain, including immune evasion by SARS-CoV-2 variants, waning of immune response, inadequate knowledge of correlates of protection, and dosing in special populations. This review serves as a detailed evaluation of the development of the current SARS-CoV-2 vaccines, their effectiveness, and challenges to their deployment as a preventive tool.
Collapse
Affiliation(s)
- Akira A Shishido
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland.,Division of Infectious Diseases, Virginia Commonwealth University, Richmond, Virginia
| | - Ashley H Barnes
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shivakumar Narayanan
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joel V Chua
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Debie Y, Van Audenaerde JRM, Vandamme T, Croes L, Teuwen LA, Verbruggen L, Vanhoutte G, Marcq E, Verheggen L, Le Blon D, Peeters B, Goossens ME, Pannus P, Ariën KK, Anguille S, Janssens A, Prenen H, Smits ELJ, Vulsteke C, Lion E, Peeters M, van Dam PA. Humoral and Cellular Immune Responses against SARS-CoV-2 after Third Dose BNT162b2 following Double-Dose Vaccination with BNT162b2 versus ChAdOx1 in Patients with Cancer. Clin Cancer Res 2023; 29:635-646. [PMID: 36341493 DOI: 10.1158/1078-0432.ccr-22-2185] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/14/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE Patients with cancer display reduced humoral responses after double-dose COVID-19 vaccination, whereas their cellular response is more comparable with that in healthy individuals. Recent studies demonstrated that a third vaccination dose boosts these immune responses, both in healthy people and patients with cancer. Because of the availability of many different COVID-19 vaccines, many people have been boosted with a different vaccine from the one used for double-dose vaccination. Data on such alternative vaccination schedules are scarce. This prospective study compares a third dose of BNT162b2 after double-dose BNT162b2 (homologous) versus ChAdOx1 (heterologous) vaccination in patients with cancer. EXPERIMENTAL DESIGN A total of 442 subjects (315 patients and 127 healthy) received a third dose of BNT162b2 (230 homologous vs. 212 heterologous). Vaccine-induced adverse events (AE) were captured up to 7 days after vaccination. Humoral immunity was assessed by SARS-CoV-2 anti-S1 IgG antibody levels and SARS-CoV-2 50% neutralization titers (NT50) against Wuhan and BA.1 Omicron strains. Cellular immunity was examined by analyzing CD4+ and CD8+ T-cell responses against SARS-CoV-2-specific S1 and S2 peptides. RESULTS Local AEs were more common after heterologous boosting. SARS-CoV-2 anti-S1 IgG antibody levels did not differ significantly between homologous and heterologous boosted subjects [GMT 1,755.90 BAU/mL (95% CI, 1,276.95-2,414.48) vs. 1,495.82 BAU/mL (95% CI, 1,131.48-1,977.46)]. However, homologous-boosted subjects show significantly higher NT50 values against BA.1 Omicron. Subjects receiving heterologous boosting demonstrated increased spike-specific CD8+ T cells, including higher IFNγ and TNFα levels. CONCLUSIONS In patients with cancer who received double-dose ChAdOx1, a third heterologous dose of BNT162b2 was able to close the gap in antibody response.
Collapse
Affiliation(s)
- Yana Debie
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium.,Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Jonas R M Van Audenaerde
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Timon Vandamme
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium.,Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Lieselot Croes
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium.,GeIntegreerd Kankercentrum Gent (IKG), AZ Maria Middelares, Gent, Belgium
| | - Laure-Anne Teuwen
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium
| | - Lise Verbruggen
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium
| | - Greetje Vanhoutte
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium
| | - Elly Marcq
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Lisa Verheggen
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium
| | - Debbie Le Blon
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Bart Peeters
- Department of Laboratory Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Maria E Goossens
- SD Infectious Diseases in Humans, Service Immune response, Sciensano, Brussels, Belgium
| | - Pieter Pannus
- SD Infectious Diseases in Humans, Service Immune response, Sciensano, Brussels, Belgium
| | - Kevin K Ariën
- Virology Unit, Institute of Tropical Medicine Antwerp (ITM), Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology (LEH), Vaxinfectio, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.,Division of Hematology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Annelies Janssens
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium.,Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Hans Prenen
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium.,Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Evelien L J Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Christof Vulsteke
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium.,GeIntegreerd Kankercentrum Gent (IKG), AZ Maria Middelares, Gent, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology (LEH), Vaxinfectio, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Marc Peeters
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium.,Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Peter A van Dam
- Multidisciplinary Oncological Center Antwerp (MOCA), Antwerp University Hospital (UZA), Edegem, Belgium.,Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
13
|
Touizer E, Alrubayyi A, Ford R, Hussain N, Gerber PP, Shum HL, Rees-Spear C, Muir L, Gea-Mallorquí E, Kopycinski J, Jankovic D, Jeffery-Smith A, Pinder CL, Fox TA, Williams I, Mullender C, Maan I, Waters L, Johnson M, Madge S, Youle M, Barber TJ, Burns F, Kinloch S, Rowland-Jones S, Gilson R, Matheson NJ, Morris E, Peppa D, McCoy LE. Attenuated humoral responses in HIV after SARS-CoV-2 vaccination linked to B cell defects and altered immune profiles. iScience 2023; 26:105862. [PMID: 36590902 PMCID: PMC9788849 DOI: 10.1016/j.isci.2022.105862] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
We assessed a cohort of people living with human immunodeficiency virus (PLWH) (n = 110) and HIV negative controls (n = 64) after 1, 2 or 3 SARS-CoV-2 vaccine doses. At all timepoints, PLWH had significantly lower neutralizing antibody (nAb) titers than HIV-negative controls. We also observed a delayed development of neutralization in PLWH that was underpinned by a reduced frequency of spike-specific memory B cells (MBCs). Improved neutralization breadth was seen against the Omicron variant (BA.1) after the third vaccine dose in PLWH but lower nAb responses persisted and were associated with global MBC dysfunction. In contrast, SARS-CoV-2 vaccination induced robust T cell responses that cross-recognized variants in PLWH. Strikingly, individuals with low or absent neutralization had detectable functional T cell responses. These PLWH had reduced numbers of circulating T follicular helper cells and an enriched population of CXCR3+CD127+CD8+T cells after two doses of SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Emma Touizer
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Aljawharah Alrubayyi
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rosemarie Ford
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Noshin Hussain
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hiu-Long Shum
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Chloe Rees-Spear
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Luke Muir
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | | | - Jakub Kopycinski
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Dylan Jankovic
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Anna Jeffery-Smith
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Christopher L. Pinder
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Thomas A. Fox
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Ian Williams
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
| | - Claire Mullender
- Institute for Global Health, University College London, London, UK
| | - Irfaan Maan
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- Institute for Global Health, University College London, London, UK
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
| | - Margaret Johnson
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Sara Madge
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Michael Youle
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Tristan J. Barber
- Institute for Global Health, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Fiona Burns
- Institute for Global Health, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Sabine Kinloch
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | | | - Richard Gilson
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- Institute for Global Health, University College London, London, UK
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Emma Morris
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Dimitra Peppa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Laura E. McCoy
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
14
|
Mihaylova A, Lesichkova S, Baleva M, Nikolova‐Vlahova M, Kundurzhiev T, Kolevski A, Naumova E. Durability of humoral and cell-mediated immune response after SARS-CoV-2 mRNA vaccine administration. J Med Virol 2023; 95:e28360. [PMID: 36448089 PMCID: PMC9878094 DOI: 10.1002/jmv.28360] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/17/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022]
Abstract
Vaccination against the SARS-Cov-2 virus is an effective way to protect against the disease and the severe course of COVID-19. Forty-nine fully vaccinated with mRNA vaccines (BNT162b2 or mRNA-1273) SARS-CoV-2 infection-naïve volunteers aged 33-89 were enrolled in the study. Evaluation of the cellular and humoral immune response was performed within 1 to 3 months (T1) and 6-9 months (T2) after the second injection, and within 2-3 months (T3) after a booster dose. Additionally, a comparative analysis of the specific immune status was made between two age groups-below 60 (n = 22) and over 60 (n = 27) years. SARS-CoV-2-specific T-cell response was evaluated by IFN-γ-producing spot forming cells (SFCs) using a standardized ELISPOT assay. Virus neutralizing antibodies (VNA) against SARS-CoV-2 were measured by a blocking ELISA test and spike protein specific IgG (S-IgG) and IgA (S-IgA) antibodies-by semiquantitative ELISA. IFN-γ-producing SFCs, S-IgG, S-IgA and VNA significantly decreased 6-9 months after the second dose. After the third injection S-IgG and S-IgA markedly increased compared to T2 and reached the levels at T1. Of note, the highest values of VNA were observed at T3. No differences in the tested immune parameters were found between the two age groups. Data obtained showed that for a long period-6-9 months after a full course of immunization with mRNA vaccine, immune reactivity is present, but both cellular and humoral immune responses gradually decrease. The administration of a third dose mainly restores the specific humoral immune response against the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Anastasiya Mihaylova
- Clinic of Clinical Immunology and Stem Cell BankUniversity Hospital AlexandrovskaSofiaBulgaria
| | - Spaska Lesichkova
- Clinic of Clinical Immunology and Stem Cell BankUniversity Hospital AlexandrovskaSofiaBulgaria
- Department of Clinical ImmunologyMedical UniversitySofiaBulgaria
| | | | - Milena Nikolova‐Vlahova
- Clinic of NephrologyUniversity Hospital St.Ivan RilskiSofiaBulgaria
- Department of Internal MedicineMedical UniversitySofiaBulgaria
| | - Todor Kundurzhiev
- Department of Occupational Medicine, Biostatistics and Medical InformaticsMedical UniversitySofiaBulgaria
| | - Alexander Kolevski
- Laboratory of MicrobiologyUniversity Hospital AlexandrovskaSofiaBulgaria
| | - Elissaveta Naumova
- Clinic of Clinical Immunology and Stem Cell BankUniversity Hospital AlexandrovskaSofiaBulgaria
- Department of Clinical ImmunologyMedical UniversitySofiaBulgaria
| |
Collapse
|
15
|
Stirrup O, Krutikov M, Tut G, Palmer T, Bone D, Bruton R, Fuller C, Azmi B, Lancaster T, Sylla P, Kaur N, Spalkova E, Bentley C, Amin U, Jadir A, Hulme S, Giddings R, Nacer-Laidi H, Baynton V, Irwin-Singer A, Hayward A, Moss P, Copas A, Shallcross L. Severe Acute Respiratory Syndrome Coronavirus 2 Anti-Spike Antibody Levels Following Second Dose of ChAdOx1 nCov-19 or BNT162b2 Vaccine in Residents of Long-term Care Facilities in England (VIVALDI). J Infect Dis 2022; 226:1877-1881. [PMID: 35429382 PMCID: PMC9047242 DOI: 10.1093/infdis/jiac146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/14/2022] [Indexed: 12/31/2022] Open
Abstract
General population studies have shown strong humoral response following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination with subsequent waning of anti-spike antibody levels. Vaccine-induced immune responses are often attenuated in frail and older populations, but published data are scarce. We measured SARS-CoV-2 anti-spike antibody levels in long-term care facility residents and staff following a second vaccination dose with Oxford-AstraZeneca or Pfizer-BioNTech. Vaccination elicited robust antibody responses in older residents, suggesting comparable levels of vaccine-induced immunity to that in the general population. Antibody levels are higher after Pfizer-BioNTech vaccination but fall more rapidly compared to Oxford-AstraZeneca recipients and are enhanced by prior infection in both groups.
Collapse
Affiliation(s)
- Oliver Stirrup
- Institute for Global Health, University College London, United Kingdom
| | - Maria Krutikov
- Institute of Health Informatics, University College London, United Kingdom
| | - Gokhan Tut
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Tom Palmer
- Institute for Global Health, University College London, United Kingdom
| | - David Bone
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Rachel Bruton
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Chris Fuller
- Institute of Health Informatics, University College London, United Kingdom
| | - Borscha Azmi
- Institute of Health Informatics, University College London, United Kingdom
| | - Tara Lancaster
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Panagiota Sylla
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Nayandeep Kaur
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Eliska Spalkova
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Christopher Bentley
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Umayr Amin
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Azar Jadir
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Samuel Hulme
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Rebecca Giddings
- Institute of Health Informatics, University College London, United Kingdom
| | - Hadjer Nacer-Laidi
- Institute of Health Informatics, University College London, United Kingdom
| | - Verity Baynton
- Department of Health and Social Care, London, United Kingdom
| | | | - Andrew Hayward
- Institute of Epidemiology and Health Care, University College London, United Kingdom
- Health Data Research UK, London, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, United Kingdom
| | - Andrew Copas
- Institute for Global Health, University College London, United Kingdom
| | - Laura Shallcross
- Institute of Health Informatics, University College London, United Kingdom
| |
Collapse
|
16
|
Stolfi P, Castiglione F, Mastrostefano E, Di Biase I, Di Biase S, Palmieri G, Prisco A. In-silico evaluation of adenoviral COVID-19 vaccination protocols: Assessment of immunological memory up to 6 months after the third dose. Front Immunol 2022; 13:998262. [PMID: 36353634 PMCID: PMC9639861 DOI: 10.3389/fimmu.2022.998262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/22/2022] [Indexed: 11/25/2022] Open
Abstract
Background The immune response to adenoviral COVID-19 vaccines is affected by the interval between doses. The optimal interval is unknown. Aim We aim to explore in-silico the effect of the interval between vaccine administrations on immunogenicity and to analyze the contribution of pre-existing levels of antibodies, plasma cells, and memory B and T lymphocytes. Methods We used a stochastic agent-based immune simulation platform to simulate two-dose and three-dose vaccination protocols with an adenoviral vaccine. We identified the model's parameters fitting anti-Spike antibody levels from individuals immunized with the COVID-19 vaccine AstraZeneca (ChAdOx1-S, Vaxzevria). We used several statistical methods, such as principal component analysis and binary classification, to analyze the correlation between pre-existing levels of antibodies, plasma cells, and memory B and T cells to the magnitude of the antibody response following a booster dose. Results and conclusions We find that the magnitude of the antibody response to a booster depends on the number of pre-existing memory B cells, which, in turn, is highly correlated to the number of T helper cells and plasma cells, and the antibody titers. Pre-existing memory T cytotoxic cells and antibodies directly influence antigen availability hence limiting the magnitude of the immune response. The optimal immunogenicity of the third dose is achieved over a large time window, spanning from 6 to 16 months after the second dose. Interestingly, after any vaccine dose, individuals can be classified into two groups, sustainers and decayers, that differ in the kinetics of decline of their antibody titers due to differences in long-lived plasma cells. This suggests that the decayers may benefit from a tailored boosting schedule with a shorter interval to avoid the temporary loss of serological immunity.
Collapse
Affiliation(s)
- Paola Stolfi
- Institute for Applied Computing, National Research Council of Italy, Rome, Italy
| | - Filippo Castiglione
- Institute for Applied Computing, National Research Council of Italy, Rome, Italy
| | - Enrico Mastrostefano
- Institute for Applied Computing, National Research Council of Italy, Rome, Italy
| | | | | | - Gianna Palmieri
- Institute of Biosciences and BioResources, National Research Council, Naples, Italy
| | - Antonella Prisco
- Institute of Genetics and Biophysics, National Research Council, Naples, Italy
| |
Collapse
|
17
|
Aviv A. The bullwhip effect, T-cell telomeres, and SARS-CoV-2. THE LANCET. HEALTHY LONGEVITY 2022; 3:e715-e721. [PMID: 36202131 PMCID: PMC9529217 DOI: 10.1016/s2666-7568(22)00190-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 01/15/2023] Open
Abstract
Both myeloid cells, which contribute to innate immunity, and lymphoid cells, which dominate adaptive immunity, partake in defending against SARS-CoV-2. In response to the virus, the otherwise slow haematopoietic production supply chain quickly unleashes its preconfigured myeloid element, which largely resists a bullwhip-like effect. By contrast, the lymphoid element risks a bullwhip-like effect when it produces T cells and B cells that are specifically designed to clear the virus. As T-cell production is telomere-length dependent and telomeres shorten with age, older adults are at higher risk of a T-cell shortfall when contracting SARS-CoV-2 than are younger adults. A poorly calibrated adaptive immune response, stemming from a bullwhip-like effect, compounded by a T-cell deficit, might thus contribute to the propensity of people with inherently short T-cell telomeres to develop severe COVID-19. The immune systems of these individuals might also generate an inadequate T-cell response to anti-SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Abraham Aviv
- Center of Human Development and Aging, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|
18
|
Muacevic A, Adler JR. COVID-19 Vaccination a Cause of Guillain-Barré Syndrome? A Case Series. Cureus 2022; 14:e30888. [PMID: 36465787 PMCID: PMC9709351 DOI: 10.7759/cureus.30888] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 01/25/2023] Open
Abstract
Guillain-Barré syndrome (GBS) is a rare autoimmune neuropathic disorder of peripheral nerves usually following an infection or on rarer occasions following vaccinations, but the exact underlying pathophysiology is still unclear. The most common etiology of GBS is a bacterial infection caused by Campylobacter jejuni. Viral infections like Zika virus, Epstein-Barr virus, and Cytomegalovirus also add to the list of GBS etiology. COVID-19 (SARS-CoV-2) has also been reported to cause GBS. Vaccines like the rabies vaccine, influenza vaccine, and poliovirus vaccine account for a very small fraction of Guillain-Barré syndrome. GBS as an adverse effect of COVID-19 vaccination was not reported by the Vaccine Adverse Event Reporting System (VAERS), but an update was later released in the course of the pandemic from FDA news, reporting several patients developing GBS after receiving the COVID-19 vaccine. In this case series, we discuss five cases that developed the GBS post-COVID-19 AstraZeneca vaccine, along with its pathophysiology, management, and outcome.
Collapse
|
19
|
Parry H, Bruton R, Ayodele R, Sylla P, McIlroy G, Logan N, Scott S, Nicol S, Verma K, Stephens C, Willett B, Zuo J, Moss P. Vaccine subtype and dose interval determine immunogenicity of primary series COVID-19 vaccines in older people. Cell Rep Med 2022; 3:100739. [PMID: 36075216 PMCID: PMC9404227 DOI: 10.1016/j.xcrm.2022.100739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/29/2022] [Accepted: 08/18/2022] [Indexed: 01/02/2023]
Abstract
Age is the strongest determinant of COVID-19 mortality, and over 2 billion people have received primary series vaccination with BNT162b2 (mRNA) or ChAdOx1 (adenoviral vector). However, the profile of sustained vaccine immunogenicity in older people is unknown. Here, we determine spike-specific humoral and cellular immunity to 8 months following BNT162b2 or ChAdOx1 in 245 people aged 80-98 years. Vaccines are strongly immunogenic, with antibodies retained in every donor, while titers fall to 23%-26% from peak. Peak immunity develops rapidly with standard interval BNT162b2, although antibody titers are enhanced 3.7-fold with extended interval. Neutralization of ancestral variants is superior following BNT162b2, while neutralization of Omicron is broadly negative. Conversely, cellular responses are stronger following ChAdOx1 and are retained to 33%-60% of peak with all vaccines. BNT162b2 and ChAdOx1 elicit strong, but differential, sustained immunogenicity in older people. These data provide a baseline to assess optimal booster regimen in this vulnerable age group.
Collapse
Affiliation(s)
- Helen Parry
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Rachel Bruton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Reni Ayodele
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Penny Sylla
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Graham McIlroy
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Nicola Logan
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G61 1QH, UK
| | - Sam Scott
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G61 1QH, UK
| | - Sam Nicol
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Kriti Verma
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Christine Stephens
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Brian Willett
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow G61 1QH, UK
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
20
|
Alharbi NK, Al-Tawfiq JA, Alwehaibe A, Alenazi MW, Almasoud A, Algaisi A, Alhumaydhi FA, Hashem AM, Bosaeed M, Alsagaby SA. Persistence of Anti-SARS-CoV-2 Spike IgG Antibodies Following COVID-19 Vaccines. Infect Drug Resist 2022; 15:4127-4136. [PMID: 35937784 PMCID: PMC9348632 DOI: 10.2147/idr.s362848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/27/2022] [Indexed: 11/26/2022] Open
Abstract
Purpose This study was conducted to investigate antibody immune responses induced by BNT162b2 and AZD1222 human COVID-19 vaccines in Riyadh city, Saudi Arabia. Patients and Methods ELISA was used to evaluate antibodies, against the SARS-CoV-2 spike S1 protein, in serum samples from 432 vaccinated individuals at six time points: pre-vaccination (baseline), post-prime, post-boost, 6-months, and 1 year post-vaccination, and 3 weeks post a third dose. Virus microneutralization assay was used to confirm antibody responses in a subset of samples. Results Anti-SARS-CoV-2 spike IgG were detected in most subjects post-prime, reached a peak level post-boost, and remained at high level at the 6-month follow-up. At 1 year post-vaccine, the antibody levels were low but increased to a significant level higher than the peak following a third dose. The third dose was given at an average of 250 days after the second dose. The virus microneutralization assay confirmed the neutralization activity of the induced SARS-CoV-2 IgG antibodies. The vaccines induced higher IgG titres at post-prime (p=0.0001) and 6 months (p=0.006) in previously infected individuals. An increased interval between prime and boost, more than recommended time, appeared to enhance the IgG levels (p=0004). Moreover, the vaccines induced higher IgG levels in younger subjects (p=0.01). Conclusion These data provide insights and build on the current understanding of immune responses induced by these two vaccines; and support a third boosting dose for these COVID-19 vaccines.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- Vaccine Development Unit, King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
- Correspondence: Naif Khalaf Alharbi, Email
| | - Jaffar A Al-Tawfiq
- Specialty Internal Medicine and Quality Department, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- Infectious Diseases Division, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Infectious Diseases Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amal Alwehaibe
- Vaccine Development Unit, King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Mohamed W Alenazi
- Vaccine Development Unit, King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Abdullah Algaisi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Anwar M Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Bosaeed
- Vaccine Development Unit, King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Science (KSAU-HS), Riyadh, Saudi Arabia
- King Abdulaziz Medical City (KAMC), Ministry of National Guard – Health Affairs (MNG-HA), Riyadh, Saudi Arabia
| | - Suliman A Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| |
Collapse
|
21
|
Caimi B, Franzetti M, Velleca R, Lai A, Gatti A, Rossi PL, D'Orso M, Pregliasco F, Balotta C, Calicchio G. Sero-survey on long-term care facility residents reveals increased risk of sub-optimal antibody response to BNT162b2: implications for breakthrough prevention. BMC Geriatr 2022; 22:191. [PMID: 35272615 PMCID: PMC8908300 DOI: 10.1186/s12877-022-02884-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/09/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The impact of coronavirus disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) on residents of long-term care facilities (LTCFs) has been dramatic on global scale as older age and comorbidities pose an increased risk of severe disease and death. METHODS Aim of this study was to evaluate SARS-CoV-2 Spike-specific IgG (S-IgG) antibody titers in 478 residents and 649 health care workers of a large Italian long-term care facility two months after complete vaccination with BNT162b2. Associations among resident-related factors and predictors of humoral response were investigated. RESULTS By stratifying levels of humoral responses, we found that 62.1%, 21.6%, 12.1% and 4.2% of residents had high (>1,000 BAU/ml), medium (101-1,000), low (1-100) and null (<1 BAU/mL) S-IgG titers, respectively. Residents with documented previous COVID-19 and those with SARS-CoV-2 nucleocapsid-specific IgG (N-IgG) positive serology showed higher level of serological response, while significant associations were observed for cancer with suboptimal response (p = 0.005) and the administration of corticosteroid for suboptimal response (p = 0.028) and a null one (p = 0.039). According to multivariate logistic regression, predictors of an increased risk of null response were advanced age (Odd ratio, OR: 2.630; Confidence interval, CI: 1.13-6.14; p = 0.025), corticosteroid therapy (OR: 4.964; CI: 1.06-23.52; p = 0.042) and diabetes mellitus (OR:3.415; CI:1.08-10.8; p = 0.037). In contrast, previous diagnosis of COVID-19 was strongly associated with a reduced risk of null response to vaccination (OR:0.126; CI:0.02-0.23; p < 0.001). CONCLUSIONS SARS-CoV-2 specific antibodies in elderly individuals should be consider when deciding the need of a third dose of vaccine for prevention of reinfections in LTCFs despite the maintenance of barrier measures.
Collapse
Affiliation(s)
- Barbara Caimi
- Azienda Servizi alla Persona, Istituti Milanesi Martinitt e Stelline e Pio Albergo Trivulzio, Milan, Italy
| | - Marco Franzetti
- Infectious Diseases Unit, Legnano General Hospital, ASST Ovest Milanese, Legnano, Italy
| | - Rossella Velleca
- Azienda Servizi alla Persona, Istituti Milanesi Martinitt e Stelline e Pio Albergo Trivulzio, Milan, Italy
| | - Alessia Lai
- Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Via G.B. Grassi, 74, 20157, Milan, Italy.
| | - Antonella Gatti
- Azienda Servizi alla Persona, Istituti Milanesi Martinitt e Stelline e Pio Albergo Trivulzio, Milan, Italy
| | - Pier Luigi Rossi
- Azienda Servizi alla Persona, Istituti Milanesi Martinitt e Stelline e Pio Albergo Trivulzio, Milan, Italy
| | - Marco D'Orso
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Fabrizio Pregliasco
- Department of Biomedical Sciences, University of Milan and IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Claudia Balotta
- Department of Biomedical and Clinical Sciences L. Sacco, University of Milan, Via G.B. Grassi, 74, 20157, Milan, Italy
| | - Giuseppe Calicchio
- Azienda Servizi alla Persona, Istituti Milanesi Martinitt e Stelline e Pio Albergo Trivulzio, Milan, Italy
| |
Collapse
|
22
|
Saleem B, Ross RL, Duquenne L, Hughes P, Emery P. COVID-19 vaccine-induced T-cell responses in patients with rheumatoid arthritis: preferential induction by ChAdOx1. THE LANCET RHEUMATOLOGY 2022; 4:e171-e172. [PMID: 35136863 PMCID: PMC8813059 DOI: 10.1016/s2665-9913(22)00027-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
23
|
Abstract
The adaptive immune response is a major determinant of the clinical outcome after SARS-CoV-2 infection and underpins vaccine efficacy. T cell responses develop early and correlate with protection but are relatively impaired in severe disease and are associated with intense activation and lymphopenia. A subset of T cells primed against seasonal coronaviruses cross reacts with SARS-CoV-2 and may contribute to clinical protection, particularly in early life. T cell memory encompasses broad recognition of viral proteins, estimated at around 30 epitopes within each individual, and seems to be well sustained so far. This breadth of recognition can limit the impact of individual viral mutations and is likely to underpin protection against severe disease from viral variants, including Omicron. Current COVID-19 vaccines elicit robust T cell responses that likely contribute to remarkable protection against hospitalization or death, and novel or heterologous regimens offer the potential to further enhance cellular responses. T cell immunity plays a central role in the control of SARS-CoV-2 and its importance may have been relatively underestimated thus far.
Collapse
Affiliation(s)
- Paul Moss
- University of Birmingham, Birmingham, UK.
| |
Collapse
|
24
|
Extended interval BNT162b2 vaccination enhances peak antibody generation. NPJ Vaccines 2022; 7:14. [PMID: 35087066 PMCID: PMC8795435 DOI: 10.1038/s41541-022-00432-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
The BNT162b2 vaccine is highly effective against COVID-19 infection and was delivered with a 3-week time interval in registration studies1. However, many countries extended this interval to accelerate population coverage with a single vaccine. It is not known how immune responses are influenced by delaying the second dose. We provide the assessment of immune responses in the first 14 weeks after standard or extended-interval BNT162b2 vaccination and show that delaying the second dose strongly boosts the peak antibody response by 3.5-fold in older people. This enhanced antibody response may offer a longer period of clinical protection and delay the need for booster vaccination. In contrast, peak cellular-specific responses were the strongest in those vaccinated on a standard 3-week vaccine interval. As such, the timing of the second dose has a marked influence on the kinetics and magnitude of the adaptive immune response after mRNA vaccination in older people.
Collapse
|
25
|
Chen CY, Liu KT, Shih SR, Ye JJ, Chen YT, Pan HC, Hsu HJ, Sun CY, Lee CC, Wu CY, Lai CC, Wu IW. Neutralization Assessments Reveal High Cardiothoracic Ratio and Old Age as Independent Predictors of Low Neutralizing Antibody Titers in Hemodialysis Patients Receiving a Single Dose of COVID-19 Vaccine. J Pers Med 2022; 12:68. [PMID: 35055386 PMCID: PMC8781271 DOI: 10.3390/jpm12010068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Data are lacking regarding predictors of quantification of neutralizing antibodies (nAbs) based on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) 50% neutralization titer (NT50) after a single dose of COVID-19 vaccine in hemodialysis (HD) patients. METHODS This prospective single-center study enrolled 200 HD patients and 82 healthy subjects to estimate antibodies against the SARS-CoV-2 viral spike protein 1 and receptor-binding domain after a first dose of a COVID-19 vaccine (ChAdOx1 or mRNA-1273), measured by enzyme-linked immunosorbent assay and applied spline-based generalized additive model regression analysis to predict NT50 converted to international units. RESULTS After the first dose of ChAdOx1, multiple linear regression showed that age (p = 0.011) and cardiothoracic ratio (p = 0.002) were negatively associated with NT50. Older age (OR = 0.958, p = 0.052) and higher cardiothoracic ratio (OR < 0.001, p = 0.037) could predict negative humoral response (NT50 < 35.13 IU/mL). NT50 was lower in HD patients compared with healthy controls receiving ChAdOx1 (10.68 vs. 43.01 IU/m, p < 0.001) or mRNA-1273 (36.39 vs. 262.2 IU/mL, p < 0.001). ChAdOx1 elicited lower GMTs than mRNA-1273 in the HD cohort (10.68 vs. 36.39 IU/mL, p < 0.001) and in healthy controls (43.01 vs. 262.22 IU/mL, p < 0.001). CONCLUSION High cardiothoracic ratio and old age could independently predict a decline in nAb titers in an HD cohort vaccinated with a single dose of ChAdOx1.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-Y.C.); (Y.-T.C.); (H.-C.P.); (H.-J.H.); (C.-Y.S.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Kuan-Ting Liu
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (K.-T.L.); (S.-R.S.)
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (K.-T.L.); (S.-R.S.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Jung-Jr Ye
- Department of Infectious Diseases, Chang Gung Memorial Hospital, Keelung 204, Taiwan;
| | - Yih-Ting Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-Y.C.); (Y.-T.C.); (H.-C.P.); (H.-J.H.); (C.-Y.S.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Heng-Chih Pan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-Y.C.); (Y.-T.C.); (H.-C.P.); (H.-J.H.); (C.-Y.S.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Heng-Jung Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-Y.C.); (Y.-T.C.); (H.-C.P.); (H.-J.H.); (C.-Y.S.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Chiao-Yin Sun
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-Y.C.); (Y.-T.C.); (H.-C.P.); (H.-J.H.); (C.-Y.S.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Chin-Chan Lee
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-Y.C.); (Y.-T.C.); (H.-C.P.); (H.-J.H.); (C.-Y.S.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Chun-Ying Wu
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan;
| | - Chi-Chun Lai
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - I-Wen Wu
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (C.-Y.C.); (Y.-T.C.); (H.-C.P.); (H.-J.H.); (C.-Y.S.); (C.-C.L.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| |
Collapse
|
26
|
COVID-19 mRNA Based Vaccine Immune-Response Assessment in Nursing Home Residents for Public Health Decision. Vaccines (Basel) 2021; 9:vaccines9121429. [PMID: 34960173 PMCID: PMC8703754 DOI: 10.3390/vaccines9121429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
Nursing home residents (NHR) have been targeted as a vaccination priority due to their higher risk of worse outcome after COVID-19 infection. The mRNA-based vaccine BTN2b2 was first approved in Europe for NHRs. The assessment of the specific vaccine immune response (both humoral and cellular) at long term in NHRs has not been addressed yet. A representative sample of 624 NHR subjects in Northern region of Spain was studied to assess immune response against full vaccination with BTN2b2. The anti-S1 antibody levels and specific T cells were measured at two and six months after vaccination. 24.4% of NHR had a previous infection prior to vaccination. The remaining NHR were included in the full vaccination assessment group (FVA). After two months, a 94.9% of the FVA presented anti-S1 antibodies, whereas those seronegative without specific cellular response were 2.54%. At long-term, the frequency of NHR within the FVA group with anti-S1 antibodies at six months were 88.12% and the seronegative subjects without specific cellular response was 8.07%. The cellular immune assays complement the humoral test in the immune vaccine response assessment. Therefore, the cellular immune assessment in NHRs allows for the fine tuning of those seronegative subjects with potential competent immune responses against the vaccine.
Collapse
|
27
|
Jeffery-Smith A, Rowland TAJ, Patel M, Whitaker H, Iyanger N, Williams SV, Giddings R, Thompson L, Zavala M, Aiano F, Ellis J, Lackenby A, Höschler K, Brown K, Ramsay ME, Gopal R, Chow JY, Ladhani SN, Zambon M. Reinfection with new variants of SARS-CoV-2 after natural infection: a prospective observational cohort in 13 care homes in England. THE LANCET HEALTHY LONGEVITY 2021; 2:e811-e819. [PMID: 34873592 PMCID: PMC8635459 DOI: 10.1016/s2666-7568(21)00253-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Understanding the duration of protection and risk of reinfection after natural infection is crucial to planning COVID-19 vaccination for at-risk groups, including care home residents, particularly with the emergence of more transmissible variants. We report on the duration, neutralising activity, and protection against the alpha variant of previous SARS-CoV-2 infection in care home residents and staff infected more than 6 months previously. Methods We did this prospective observational cohort surveillance in 13 care homes in Greater London, England. All staff and residents were included. Staff and residents had regular nose and throat screening for SARS-CoV-2 by RT-PCR according to national guidelines, with ad hoc testing of symptomatic individuals. From January, 2021, antigen lateral flow devices were also used, but positive tests still required RT-PCR confirmation. Staff members took the swab samples for themselves and the residents. The primary outcome was SARS-CoV-2 RT-PCR positive primary infection or reinfection in previously infected individuals, as determined by previous serological testing and screening or diagnostic RT-PCR results. Poisson regression and Cox proportional hazards models were used to estimate protective effectiveness of previous exposure. SARS-CoV-2 spike, nucleoprotein, and neutralising antibodies were assessed at multiple timepoints as part of the longitudinal follow-up. Findings Between April 10 and Aug 3, 2020, we recruited and tested 1625 individuals (933 staff and 692 residents). 248 participants were lost to follow-up (123 staff and 125 residents) and 1377 participants were included in the follow-up period to Jan 31, 2021 (810 staff and 567 residents). There were 23 reinfections (ten confirmed, eight probable, five possible) in 656 previously infected individuals (366 staff and 290 residents), compared with 165 primary infections in 721 susceptible individuals (444 staff and 277 residents). Those with confirmed reinfections had no or low neutralising antibody concentration before reinfection, with boosting of titres after reinfection. Kinetics of binding and neutralising antibodies were similar in older residents and younger staff. Interpretation SARS-CoV-2 reinfections were rare in older residents and younger staff. Protection from SARS-CoV-2 was sustained for longer than 9 months, including against the alpha variant. Reinfection was associated with no or low neutralising antibody before reinfection, but significant boosting occurred on reinfection. Funding Public Health England.
Collapse
Affiliation(s)
- Anna Jeffery-Smith
- Virus Reference Department, Public Health England, London, UK
- Blizard Institute, Queen Mary University of London, London, UK
- Correspondence to: Dr Anna Jeffery-Smith, Virus Reference Department, Public Health England, London NW9 5EQ, UK
| | | | - Monika Patel
- Virus Reference Department, Public Health England, London, UK
| | | | - Nalini Iyanger
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Sarah V Williams
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Rebecca Giddings
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Leah Thompson
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Maria Zavala
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Felicity Aiano
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Joanna Ellis
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Angie Lackenby
- Virus Reference Department, Public Health England, London, UK
| | - Katja Höschler
- Virus Reference Department, Public Health England, London, UK
| | - Kevin Brown
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Mary E Ramsay
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Robin Gopal
- Virus Reference Department, Public Health England, London, UK
| | - J Yimmy Chow
- London Coronavirus Response Cell, National Infection Service, Public Health England, London, UK
| | - Shamez N Ladhani
- Immunisation and Countermeasures Division, Public Health England, London, UK
- Paediatric Infectious Diseases Research Group, St. George's University of London, London, UK
| | - Maria Zambon
- Virus Reference Department, Public Health England, London, UK
| |
Collapse
|
28
|
Marfe G, Perna S, Shukla AK. Effectiveness of COVID-19 vaccines and their challenges (Review). Exp Ther Med 2021; 22:1407. [PMID: 34676000 PMCID: PMC8524740 DOI: 10.3892/etm.2021.10843] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
At the end of 2019, a new disease recognized such as severe acute respiratory syndrome (SARS), was reported in Wuhan, China. This disease was caused by an unknown SARS coronavirus 2 (SARS-CoV-2); a virus is characterized by high infectivity among humans. In some cases, this disease can be asymptomatic, while in other cases can induce flu-like symptoms or acute respiratory distress syndrome, pneumonia and death. For this reason, the World Health Organization and Public Health Emergency of International Concern declared a pandemic status in January 2020. Currently, numerous countries have been involved in the development of effective vaccines to protect humans against SARS-CoV-2 infection. The present review will discuss the four vaccines, AZD1222 (AstraZeneca or Vaxzevria), Janssen (Ad26.COV2.S), Moderna/mRNA-1273 and BioNTech/Fosun/Pfizer BNT162b1, that are currently in use worldwide to understand their efficacy, but also evaluate the difficulties and challenges of vaccine development. Although several questions should be addressed regarding these vaccines, the current review will examine the viral elements used in the coronavirus-19 vaccine that can play a crucial role in inducing a strong immune response, as well as the different adverse effects that they can cause to individuals.
Collapse
Affiliation(s)
- Gabriella Marfe
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Stefania Perna
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Arvind Kumar Shukla
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Gyeongsangnam-do 50612, Republic of Korea
- Inventra Medclin Biomedical Healthcare and Research Center, Katemanivli, Kalyan, Thane, Maharashtra 421306, India
| |
Collapse
|
29
|
Tartof SY, Slezak JM, Fischer H, Hong V, Ackerson BK, Ranasinghe ON, Frankland TB, Ogun OA, Zamparo JM, Gray S, Valluri SR, Pan K, Angulo FJ, Jodar L, McLaughlin JM. Effectiveness of mRNA BNT162b2 COVID-19 vaccine up to 6 months in a large integrated health system in the USA: a retrospective cohort study. Lancet 2021; 398:1407-1416. [PMID: 34619098 PMCID: PMC8489881 DOI: 10.1016/s0140-6736(21)02183-8] [Citation(s) in RCA: 770] [Impact Index Per Article: 256.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Vaccine effectiveness studies have not differentiated the effect of the delta (B.1.617.2) variant and potential waning immunity in observed reductions in effectiveness against SARS-CoV-2 infections. We aimed to evaluate overall and variant-specific effectiveness of BNT162b2 (tozinameran, Pfizer-BioNTech) against SARS-CoV-2 infections and COVID-19-related hospital admissions by time since vaccination among members of a large US health-care system. METHODS In this retrospective cohort study, we analysed electronic health records of individuals (≥12 years) who were members of the health-care organisation Kaiser Permanente Southern California (CA, USA), to assess BNT162b2 vaccine effectiveness against SARS-CoV-2 infections and COVID-19-related hospital admissions for up to 6 months. Participants were required to have 1 year or more previous membership of the organisation. Outcomes comprised SARS-CoV-2 PCR-positive tests and COVID-19-related hospital admissions. Effectiveness calculations were based on hazard ratios from adjusted Cox models. This study was registered with ClinicalTrials.gov, NCT04848584. FINDINGS Between Dec 14, 2020, and Aug 8, 2021, of 4 920 549 individuals assessed for eligibility, we included 3 436 957 (median age 45 years [IQR 29-61]; 1 799 395 [52·4%] female and 1 637 394 [47·6%] male). For fully vaccinated individuals, effectiveness against SARS-CoV-2 infections was 73% (95% CI 72-74) and against COVID-19-related hospital admissions was 90% (89-92). Effectiveness against infections declined from 88% (95% CI 86-89) during the first month after full vaccination to 47% (43-51) after 5 months. Among sequenced infections, vaccine effectiveness against infections of the delta variant was high during the first month after full vaccination (93% [95% CI 85-97]) but declined to 53% [39-65] after 4 months. Effectiveness against other (non-delta) variants the first month after full vaccination was also high at 97% (95% CI 95-99), but waned to 67% (45-80) at 4-5 months. Vaccine effectiveness against hospital admissions for infections with the delta variant for all ages was high overall (93% [95% CI 84-96]) up to 6 months. INTERPRETATION Our results provide support for high effectiveness of BNT162b2 against hospital admissions up until around 6 months after being fully vaccinated, even in the face of widespread dissemination of the delta variant. Reduction in vaccine effectiveness against SARS-CoV-2 infections over time is probably primarily due to waning immunity with time rather than the delta variant escaping vaccine protection. FUNDING Pfizer.
Collapse
Affiliation(s)
- Sara Y Tartof
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA; Department of Health Systems Science, Kaiser Permanente Bernard J Tyson School of Medicine, Pasadena, CA, USA.
| | - Jeff M Slezak
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Heidi Fischer
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Vennis Hong
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Bradley K Ackerson
- Department of Pediatric Infectious Diseases, Southern California Permanente Medical Group, Harbor City, CA, USA
| | - Omesh N Ranasinghe
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Timothy B Frankland
- Kaiser Permanente Center for Integrated Health Care Research, Honolulu, HI, USA
| | - Oluwaseye A Ogun
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Iversen PL, Bavari S. Extending the interval of COVID-19 vaccine regimens in individuals aged 80 years or older. THE LANCET. HEALTHY LONGEVITY 2021; 2:e529-e530. [PMID: 34430953 PMCID: PMC8376211 DOI: 10.1016/s2666-7568(21)00205-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Patrick L Iversen
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, USA
| | - Sina Bavari
- Edge BioInnovation and Healion Bio, Frederick, MD 21702, USA
| |
Collapse
|