1
|
Metko M, Tonne J, Veliz Rios A, Thompson J, Mudrick H, Masopust D, Diaz RM, Barry MA, Vile RG. Intranasal Prime-Boost with Spike Vectors Generates Antibody and T-Cell Responses at the Site of SARS-CoV-2 Infection. Vaccines (Basel) 2024; 12:1191. [PMID: 39460356 PMCID: PMC11511174 DOI: 10.3390/vaccines12101191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Long-lived, re-activatable immunity to SARS-CoV-2 and its emerging variants will rely on T cells recognizing conserved regions of viral proteins across strains. Heterologous prime-boost regimens can elicit elevated levels of circulating CD8+ T cells that provide a reservoir of first responders upon viral infection. Although most vaccines are currently delivered intramuscularly (IM), the initial site of infection is the nasal cavity. METHODS Here, we tested the hypothesis that a heterologous prime and boost vaccine regimen delivered intranasally (IN) will generate improved immune responses locally at the site of virus infection compared to intramuscular vaccine/booster regimens. RESULTS In a transgenic human ACE2 murine model, both a Spike-expressing single-cycle adenovirus (SC-Ad) and an IFNß safety-enhanced replication-competent Vesicular Stomatitis Virus (VSV) platform generated anti-Spike antibody and T-cell responses that diminished with age. Although SC-Ad-Spike boosted a prime with VSV-Spike-mIFNß, SC-Ad-Spike alone induced maximal levels of IgG, IgA, and CD8+ T-cell responses. CONCLUSIONS There were significant differences in T-cell responses in spleens compared to lungs, and the intranasal boost was significantly superior to the intramuscular boost in generating sentinel immune effectors at the site of the virus encounter in the lungs. These data show that serious consideration should be given to intranasal boosting with anti-SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Muriel Metko
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Alexa Veliz Rios
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Haley Mudrick
- Molecular Pharmacology and Experimental Therapeutics Program, Mayo Clinic, Rochester, MN 55905, USA;
| | - David Masopust
- Department of Microbiology & Immunology, University of Minnesota Medical School, 2101 6th St. SE, Minneapolis, MN 55455, USA;
| | - Rosa Maria Diaz
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
| | - Michael A. Barry
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
- Department of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard G. Vile
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (M.M.); (J.T.); (A.V.R.); (J.T.); (R.M.D.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
2
|
Page L, Dennehy K, Mueller K, Girl P, Loell E, Buijze H, Classen JM, Messmann H, Roemmele C, Hoffmann R, Wurster S, Fuchs A. Antigen-specific T helper cells and cytokine profiles predict intensity and longevity of cellular and humoral responses to SARS-CoV-2 booster vaccination. Front Immunol 2024; 15:1423766. [PMID: 39267758 PMCID: PMC11390417 DOI: 10.3389/fimmu.2024.1423766] [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: 04/26/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction Pre-existent pools of coronavirus-specific or cross-reactive T cells were shown to shape the development of cellular and humoral immune responses after primary mRNA vaccination against SARS-CoV-2. However, the cellular determinants of responses to booster vaccination remain incompletely understood. Therefore, we phenotypically and functionally characterized spike antigen-specific T helper (Th) cells in healthy, immunocompetent individuals and correlated the results with cellular and humoral immune responses to BNT162b2 booster vaccination over a six-month period. Methods Blood of 30 healthy healthcare workers was collected before, 1, 3, and 6 months after their 3rd BNT162b2 vaccination. Whole blood was stimulated with spike peptides and analyzed using flow cytometry, a 13-plex cytokine assay, and nCounter-based transcriptomics. Results Spike-specific IgG levels at 1 month after booster vaccination correlated with pre-existing CD154+CD69+IFN-γ+CD4+ effector memory cells as well as spike-induced IL-2 and IL-17A secretion. Early post-booster (1-month) spike IgG levels (r=0.49), spike-induced IL‑2 (r=0.58), and spike-induced IFN‑γ release (r=0.43) correlated moderately with their respective long-term (6-month) responses. Sustained robust IgG responses were significantly associated with S-specific (CD69+±CD154+±IFN-γ+) Th-cell frequencies before booster vaccination (p=0.038), especially double/triple-positive type-1 Th cells. Furthermore, spike IgG levels, spike-induced IL‑2 release, and spike-induced IFN‑γ release after 6 months were significantly associated with increased IL‑2 & IL‑4, IP‑10 & MCP1, and IFN‑γ & IP‑10 levels at 1 month post-booster, respectively. On the transcriptional level, induction of pathways associated with both T-cell proliferation and antigen presentation was indicative of sustained spike-induced cytokine release and spike-specific IgG production 6 months post-booster. Using support vector machine models, pre-booster spike-specific T-cell frequencies and early post-booster cytokine responses predicted sustained (6-month) responses with F1 scores of 0.80-1.00. Discussion In summary, spike-specific Th cells and T-cellular cytokine signatures present before BNT162b2 booster vaccination shape sustained adaptive cellular and humoral responses post-booster. Functional T-cell assays might facilitate early identification of potential non-responders.
Collapse
Affiliation(s)
- Lukas Page
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Kevin Dennehy
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | | | - Philipp Girl
- Bundeswehr Institute of Microbiology, Munich, Germany
- Central Institute of the Bundeswehr Medical Service, Munich, Germany
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Eva Loell
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Hellen Buijze
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Johanna-Maria Classen
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Helmut Messmann
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Christoph Roemmele
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| | - Reinhard Hoffmann
- Institute for Laboratory Medicine and Microbiology, University Hospital of Augsburg, Augsburg, Germany
| | - Sebastian Wurster
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andre Fuchs
- Internal Medicine III - Gastroenterology and Infectious Diseases, University Hospital of Augsburg, Augsburg, Germany
| |
Collapse
|
3
|
Powell AA, Dowell AC, Moss P, Ladhani SN. Current state of COVID-19 in children: 4 years on. J Infect 2024; 88:106134. [PMID: 38432584 DOI: 10.1016/j.jinf.2024.106134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
Children have been disproportionately affected by the COVID-19 pandemic. Despite evidence of a very low risk of severe disease, children were subjected to extensive lockdown, restriction and mitigation measures, including school closures, to control the rapid spread of SARS-CoV-2 in most parts of the world. In this review we summarise the UK experience of COVID-19 in children four years into the largest and longest pandemic of this century. We address the risks of SARS-CoV-2 infection, immunity, transmission, severity and outcomes in children. We also assess the implementation, uptake, effectiveness and impact of COVID-19 vaccination, as well as the emergence, evolution and near disappearance of PIMS-TS (paediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2) and current understanding of long COVID in children. This review consolidates current knowledge on childhood COVID-19 and emphasises the importance of continued research and the need for research-driven public health actions and policy decisions, especially in the context of new variants and future vaccines.
Collapse
Affiliation(s)
- Annabel A Powell
- Public Health Programmes, UK Health Security Agency, London, UK.
| | - Alexander C Dowell
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Paul Moss
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Shamez N Ladhani
- Public Health Programmes, UK Health Security Agency, London, UK; Paediatric Infectious Diseases Research Group, St. George's University of London, London, UK
| |
Collapse
|
4
|
Miyamoto S, Suzuki T. Infection-mediated immune response in SARS-CoV-2 breakthrough infection and implications for next-generation COVID-19 vaccine development. Vaccine 2024; 42:1401-1406. [PMID: 38310015 DOI: 10.1016/j.vaccine.2024.01.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
Post-vaccination infections, termed breakthrough infections, occur after the virus infection overcomes the vaccine-induced immune barrier. During the early stages of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron wave, high serum-neutralizing antibody titers against the Omicron variant were detected in individuals with breakthrough infections as well as those who received a third vaccine dose (i.e., booster recipients). Additionally, these cases indicated that Omicron antigens triggered an immune response that differed from that triggered by the vaccine strain before analysis of the effectiveness of new vaccines updated for the Omicron variants. Moreover, the magnitude and breadth of neutralizing antibody titers induced by breakthrough infections are correlated with the upper respiratory viral load at diagnosis and the duration between vaccination and infection, respectively. Unlike booster vaccine recipients, patients with breakthrough infections have varying durations between vaccination and infection. Accordingly, optimal booster vaccination intervals may be estimated based on the cross-neutralizing antibody response induced over time. Examination of breakthrough infection cases has provided valuable insights that could not be yielded by only examining vaccinated individuals alone. These insights include estimates of vaccine-induced immunity against SARS-CoV-2 variants and the various factors related to the clinical status. This review describes the immune response elicited by breakthrough infections; specifically, it discusses factors that affect the magnitude and breadth of serum antibody titers as well as the appropriate booster vaccination strategy. This review provides key aspects that could contribute to developing next-generation COVID-19 vaccines through breakthrough infection cases.
Collapse
Affiliation(s)
- Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases Tokyo 162-8640, Japan.
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases Tokyo 162-8640, Japan
| |
Collapse
|
5
|
Song Y, Lou L, Zhang K. A review of the clinical characteristics and management of immunosuppressed patients living with HIV or solid organ transplants infected with SARS-CoV-2 omicron variants. Front Public Health 2024; 12:1327093. [PMID: 38454994 PMCID: PMC10917969 DOI: 10.3389/fpubh.2024.1327093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/13/2024] [Indexed: 03/09/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) omicron strain was first detected in South Africa in November 2021. Although clinical responses to SARS-CoV-2 depend on host immunity, it remains uncertain how immunosuppression affects subsequent coronavirus disease 2019-related (COVID-19-related) incidence, severity, and mortality, especially with respect to the omicron strain. Conversely, immunosuppressants are often thought to predispose to infection. To explore the associations between host immunity and infection with SARS-CoV-2 omicron variants, here we discuss two groups of immunosuppressed patients: organ transplant recipients, who generally receive exogenous immunosuppressants, and Human Immunodeficiency Virus (HIV)-infected patients, who often have disease-related immunosuppression. In summarizing the clinical features and prognoses of HIV-infected patients and human organ transplant recipients infected with SARS-CoV-2 omicron variants, we provide new insights into the pathogenesis of omicron SARS-CoV-2 and provide a framework for the management of these patients now and in the future.
Collapse
Affiliation(s)
- Yan Song
- Department of Infectious Diseases, The First Hospital of Jilin University, Jilin, China
| | - Lixin Lou
- Department of Infectious Diseases, The First Hospital of Jilin University, Jilin, China
| | - Kaiyu Zhang
- Department of Infectious Diseases, The First Hospital of Jilin University, Jilin, China
- Department of Infectious Diseases and Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Kassianos G, MacDonald P, Aloysius I, Pather S. Responses to Common Misconceptions Relating to COVID-19 Variant-Adapted mRNA Vaccines. Vaccines (Basel) 2024; 12:57. [PMID: 38250870 PMCID: PMC10819631 DOI: 10.3390/vaccines12010057] [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: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
The evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the waning of immunity over time has necessitated the use of booster doses of original coronavirus disease 2019 (COVID-19) vaccines. This has also led to the development and implementation of variant-adapted messenger RNA (mRNA) vaccines that include an Omicron sub-lineage component in addition to the antigen based on the wild-type virus spike protein. Subsequent emergence of the recombinant XBB sub-lineages triggered the development of monovalent XBB-based variant-adapted mRNA vaccines, which are available for vaccination campaigns in late 2023. Misconceptions about new variant-adapted vaccines may exacerbate vaccine fatigue and drive the lack of vaccine acceptance. This article aims to address common concerns about the development and use of COVID-19 variant-adapted mRNA vaccines that have emerged as SARS-CoV-2 has continued to evolve.
Collapse
Affiliation(s)
- George Kassianos
- Royal College of General Practitioners, London NW1 2FB, UK;
- British Global and Travel Health Association, London NW1 2FB, UK
| | | | | | | |
Collapse
|
7
|
Madhi SA, Feikin DR. Are bivalent vaccines better than ancestral-virus monovalent vaccines in protecting against severe omicron COVID-19? THE LANCET. INFECTIOUS DISEASES 2023; 23:1325-1327. [PMID: 37478878 DOI: 10.1016/s1473-3099(23)00425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/23/2023]
Affiliation(s)
- Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Daniel R Feikin
- Department of Immunizations, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| |
Collapse
|
8
|
Bingula R, Chabrolles H, Bonnet B, Archimbaud C, Brebion A, Cosme J, Ollier A, Dutheil F, Junda M, Mirand A, Regagnon C, Vidal M, Henquell C, Evrard B. Increase over time of antibody levels 3 months after a booster dose as an indication of better protection against Omicron infection. Emerg Microbes Infect 2023; 12:2184176. [PMID: 36848040 PMCID: PMC10013501 DOI: 10.1080/22221751.2023.2184176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
The third, "booster", vaccination increases the overall immune response against SARS-CoV-2 variants. However, after the initial peak at around 3 weeks post-vaccination, anti-spike antibody levels decline. Post-booster kinetics of cellular response has been less investigated and there is no documented evidence of a true boosting effect. Furthermore, multiple studies underline the less effective immune responses against Omicron, the latest variant of concern, at both humoral and cellular levels. In this letter, we analyse humoral (anti-RBD IgG levels) and cellular (IFN-γ release assay) immune response in 205 health care workers 3 weeks and 3 months after administration of an mRNA-based booster dose, either mRNA-1273 or BNT162b2. Since all subjects were SARS-CoV-2 infection-naïve, we also looked at the incidence of Omicron infection between 3 and 6 months post-booster.At both timepoints, 3x mRNA-1273 vaccination had the highest overall antibody and IFN-γ levels, followed by 3x BNT162b2 vaccination and heterologous mRNA-based regimens. Heterologous ChAdOx1-mRNA-based regimen had the lowest antibody levels while cellular response equal to that of 3x BNT162b2 vaccination and heterologous mRNA-based regimens. Our results show that both humoral and cellular responses waned at 3 months for all vaccination regimens. However, we identified three trajectories of dosage variation. Interestingly, the subgroup of subjects with increasing anti-RBD IgG levels over time had a lower incidence of Omicron infection. Whether increasing humoral response at 3 months post-booster is more indicative of protection than a high initial peak remains to be confirmed in a larger cohort.
Collapse
Affiliation(s)
- Rea Bingula
- UMR UNH, ECREIN, Immunology Laboratory, Faculty of Medicine, Clermont Auvergne University, Clermont-Ferrand, France
| | - Hélène Chabrolles
- Virology Department, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), 3IHP, Clermont-Ferrand, France.,CNRS UMR 6023, LMGE, Clermont Auvergne University, Clermont-Ferrand, France
| | - Benjamin Bonnet
- UMR UNH, ECREIN, Immunology Laboratory, Faculty of Medicine, Clermont Auvergne University, Clermont-Ferrand, France.,Immunology Department, Clermont-Ferrand University Hospital (CHU Clermont Ferrand), Clermont-Ferrand, France
| | - Christine Archimbaud
- Virology Department, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), 3IHP, Clermont-Ferrand, France.,CNRS UMR 6023, LMGE, Clermont Auvergne University, Clermont-Ferrand, France
| | - Amélie Brebion
- Virology Department, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), 3IHP, Clermont-Ferrand, France
| | - Justine Cosme
- Immunology Department, Clermont-Ferrand University Hospital (CHU Clermont Ferrand), Clermont-Ferrand, France
| | - Amandine Ollier
- Clinical Research and Innovation Direction, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand) 3 IHP, Clermont-Ferrand, France
| | - Frédéric Dutheil
- Preventive and Occupational Medicine, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), Clermont-Ferrand, France.,CNRS, LaPSCo Physiological and Psychosocial Stress, Clermont Auvergne University, Clermont-Ferrand, France
| | - Maud Junda
- Immunology Department, Clermont-Ferrand University Hospital (CHU Clermont Ferrand), Clermont-Ferrand, France
| | - Audrey Mirand
- Virology Department, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), 3IHP, Clermont-Ferrand, France.,CNRS UMR 6023, LMGE, Clermont Auvergne University, Clermont-Ferrand, France
| | - Christel Regagnon
- Virology Department, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), 3IHP, Clermont-Ferrand, France.,CNRS UMR 6023, LMGE, Clermont Auvergne University, Clermont-Ferrand, France
| | - Magali Vidal
- Virology Department, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), 3IHP, Clermont-Ferrand, France.,CNRS UMR 6023, LMGE, Clermont Auvergne University, Clermont-Ferrand, France
| | - Cécile Henquell
- Virology Department, Clermont-Ferrand University Hospital (CHU Clermont-Ferrand), 3IHP, Clermont-Ferrand, France.,CNRS UMR 6023, LMGE, Clermont Auvergne University, Clermont-Ferrand, France
| | - Bertrand Evrard
- UMR UNH, ECREIN, Immunology Laboratory, Faculty of Medicine, Clermont Auvergne University, Clermont-Ferrand, France.,Immunology Department, Clermont-Ferrand University Hospital (CHU Clermont Ferrand), Clermont-Ferrand, France
| |
Collapse
|
9
|
Hogan AB, Wu SL, Toor J, Olivera Mesa D, Doohan P, Watson OJ, Winskill P, Charles G, Barnsley G, Riley EM, Khoury DS, Ferguson NM, Ghani AC. Long-term vaccination strategies to mitigate the impact of SARS-CoV-2 transmission: A modelling study. PLoS Med 2023; 20:e1004195. [PMID: 38016000 PMCID: PMC10715640 DOI: 10.1371/journal.pmed.1004195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 12/12/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Vaccines have reduced severe disease and death from Coronavirus Disease 2019 (COVID-19). However, with evidence of waning efficacy coupled with continued evolution of the virus, health programmes need to evaluate the requirement for regular booster doses, considering their impact and cost-effectiveness in the face of ongoing transmission and substantial infection-induced immunity. METHODS AND FINDINGS We developed a combined immunological-transmission model parameterised with data on transmissibility, severity, and vaccine effectiveness. We simulated Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) transmission and vaccine rollout in characteristic global settings with different population age-structures, contact patterns, health system capacities, prior transmission, and vaccine uptake. We quantified the impact of future vaccine booster dose strategies with both ancestral and variant-adapted vaccine products, while considering the potential future emergence of new variants with modified transmission, immune escape, and severity properties. We found that regular boosting of the oldest age group (75+) is an efficient strategy, although large numbers of hospitalisations and deaths could be averted by extending vaccination to younger age groups. In countries with low vaccine coverage and high infection-derived immunity, boosting older at-risk groups was more effective than continuing primary vaccination into younger ages in our model. Our study is limited by uncertainty in key parameters, including the long-term durability of vaccine and infection-induced immunity as well as uncertainty in the future evolution of the virus. CONCLUSIONS Our modelling suggests that regular boosting of the high-risk population remains an important tool to reduce morbidity and mortality from current and future SARS-CoV-2 variants. Our results suggest that focusing vaccination in the highest-risk cohorts will be the most efficient (and hence cost-effective) strategy to reduce morbidity and mortality.
Collapse
Affiliation(s)
- Alexandra B. Hogan
- School of Population Health, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Sean L. Wu
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, United States of America
| | - Jaspreet Toor
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Daniela Olivera Mesa
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Patrick Doohan
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Oliver J. Watson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Peter Winskill
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Giovanni Charles
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Gregory Barnsley
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Eleanor M. Riley
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David S. Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Neil M. Ferguson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| | - Azra C. Ghani
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
10
|
Yoon SW, Widyasari K, Jang J, Lee S, Kang T, Kim S. Kinetics of adaptive immune responses after administering mRNA-Based COVID-19 vaccination in individuals with and without prior SARS-CoV-2 infections. BMC Infect Dis 2023; 23:732. [PMID: 37891503 PMCID: PMC10604405 DOI: 10.1186/s12879-023-08728-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
OBJECTIVE We aimed to compare the adaptive immune response in individuals with or without prior SARS-CoV-2 infections following the administration of mRNA-based COVID-19 vaccines. METHODS A total of 54 participants with ages ranging from 37 to 56 years old, consisting of 23 individuals without a history of SARS-CoV-2 infection (uninfected group) and 31 individuals with prior infection of SARS-CoV-2 (infected group) who have received two doses of mRNA SARS-CoV-2 vaccines were enrolled in this study. We measured the IFN-γ level upon administration of BNT162b2 (PF) or mRNA-1273 (MO) by QuantiFERON SARS-CoV-2. The production of neutralizing antibodies was evaluated by a surrogate virus neutralization assay, and the neutralizing capacity was assessed by a plaque reduction neutralization test (PRNT50). The immune response was compared between the two groups. RESULTS A significantly higher level of IFN-γ (p < 0.001) and neutralization antibodies (p < 0.001) were observed in the infected group than those in the uninfected group following the first administration of vaccines. The infected group demonstrated a significantly higher PRNT50 titer than the uninfected group against the Wuhan strain (p < 0.0001). Still, the two groups were not significantly different against Delta (p = 0.07) and Omicron (p = 0.14) variants. Following the second vaccine dose, T- and B-cell levels were not significantly increased in the infected group. CONCLUSION A single dose of mRNA-based COVID-19 vaccines would boost immune responses in individuals who had previously contracted SARS-CoV-2.
Collapse
Affiliation(s)
- Sun-Woo Yoon
- Department of Biological Science and Biotechnology, Andong National University, Andong, 36729, Korea
| | - Kristin Widyasari
- Gyeongsang Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, Korea
| | - Jieun Jang
- Gyeongnam Center for Infectious Disease Control and Prevention, Changwon, 51154, Korea
| | - Seungjun Lee
- Gyeongsang Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, Korea
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, Korea
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea
| | - Sunjoo Kim
- Gyeongsang Institute of Health Sciences, Gyeongsang National University, Jinju, 52727, Korea.
- Gyeongnam Center for Infectious Disease Control and Prevention, Changwon, 51154, Korea.
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, Korea.
| |
Collapse
|
11
|
Reimann H, Moosmann C, Schober K, Lang V, Verhagen J, Zeun J, Mackensen A, Kremer AN, Völkl S, Aigner M. Identification and characterization of T-cell receptors with therapeutic potential showing conserved specificity against all SARS-CoV 2 strains. Immunobiology 2023; 228:152720. [PMID: 37541134 DOI: 10.1016/j.imbio.2023.152720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023]
Abstract
INTRODUCTION Treatment of severe COVID-19 disease can be challenging in immunocompromized patients due to newly emerging virus variants of concern (VOC) escaping the humoral response. Thus, T cells recognizing to date unmutated epitopes are not only relevant for patients' immune responses against VOC, but might also serve as a therapeutic option for patients with severe COVID-19 disease in the future, e.g. following allogenic stem cell transplantation. METHODS To this purpose, the activation, cytokine profile and specificity of T-cell clones against unmutated and omicron Spike (S)-protein was analyzed, HLA restriction was determined and most promising T-cell receptor (TCR) was introduced into allogeneic T cells via CRISPR/Cas9-mediated orthotopic TCR replacement. Finally, T-cell responses of engineered T cells was determined and durability of the TCR replacement measured. PERSPECTIVE SARS-CoV-2 specific engineered T cells recognizing a genomically stable region of the S-protein of all SARS-CoV 2 variants were successfully generated. Such transgenic T cells exhibit favorable effector functions and provide a treatment option of immunocompromised COVID-19 patients.
Collapse
Affiliation(s)
- Hannah Reimann
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany.
| | - Carolin Moosmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kilian Schober
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Vanessa Lang
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Johan Verhagen
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Department of Internal Medicine 3, Rheumatology and Immunology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Zeun
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Anita N Kremer
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
12
|
Cernera G, Gelzo M, De Placido P, Ottaviano M, Pietroluongo E, Raia M, Scalia G, Tortora M, Castaldo G, Formisano P, Palmieri G, Giuliano M. Immunocytometric analysis of patients with thymic epithelial tumors revealed that COVID-19 vaccine booster strongly enhanced the immune response. Front Immunol 2023; 14:1233056. [PMID: 37705978 PMCID: PMC10495582 DOI: 10.3389/fimmu.2023.1233056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Background Thymic epithelial tumors (TETs) are rare malignancies with heterogeneous clinical manifestations. The high frequency of autoimmune paraneoplastic disorders observed in such patients requires caution when using COVID-19 vaccines. Furthermore, TETs are often associated with severe immunodeficiency, making it difficult to predict vaccine immunization. Therefore, we aimed to evaluate immune response to COVID-19 vaccine in patients with TETs. Methods We conducted a prospective study enrolling patients who underwent the SARS-Cov-2 mRNA full vaccine cycle (two doses plus a booster after 6 months of BNT162b2). All patients were enrolled before receiving 1st vaccine dose and were followed over the vaccination cycle for up to 6 months after the booster dose to i) assess humoral and cellular responses, ii) define biomarkers predictive of effective immunization, and iii) evaluate the safety of the vaccine. Results At the end of the full vaccine cycle, 27 (61.4%) patients developed humoral and 38 (86.4%) cellular responses (IFN γ release by stimulated cells) and showed an increase in activated TH1 and TH17 cells, particularly significant after the booster dose. The number of B and T lymphocytes at baseline was predictive of humoral and cellular responses, respectively. Patients with no evidence of tumor lesions had a higher probability of achieving a humoral response than those with evidence of the disease. Furthermore, the percentage of patients with immune-related disorders (75%), particularly Good's syndrome (47.7%) and myasthenia gravis (29.5%), did not change over the entire vaccine cycle. Overall, 19 of the 44 enrolled patients (43.2%) had COVID-19 during the observation period; none required hospitalization or oxygen support, and no fatalities were observed. Conclusion SARS-Cov-2 mRNA vaccine determines the immune responses in patients with TET, particularly after the booster dose, and in patients with no evidence of tumor lesions. Preliminary analysis of B and T lymphocytes may help identify patients who have a lower probability of achieving effective humoral and cellular responses and thus may need passive immunization. The vaccine prevented severe COVID-19 infection and is safe.
Collapse
Affiliation(s)
- Gustavo Cernera
- CEINGE-Biotecnologie avanzate, scarl, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Monica Gelzo
- CEINGE-Biotecnologie avanzate, scarl, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Pietro De Placido
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - Margaret Ottaviano
- Dipartimento di Melanoma, Immunoterapia Oncologica e Terapie Innovative, IRCCS Fondazione G. Pascale, Naples, Italy
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| | - Erica Pietroluongo
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | | | | | - Marianna Tortora
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie avanzate, scarl, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, Naples, Italy
| | - Giovannella Palmieri
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| | - Mario Giuliano
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
- Centro Regionale di Coordinamento Tumori Rari Regione Campania (CRCTR), Naples, Italy
| |
Collapse
|
13
|
Song S, Madewell ZJ, Liu M, Longini IM, Yang Y. Effectiveness of SARS-CoV-2 vaccines against Omicron infection and severe events: a systematic review and meta-analysis of test-negative design studies. Front Public Health 2023; 11:1195908. [PMID: 37361171 PMCID: PMC10289159 DOI: 10.3389/fpubh.2023.1195908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Background A rapidly growing body was observed of literature evaluating the vaccine effectiveness (VE) against Omicron in test-negative design studies. Methods We systematically searched papers that evaluated VE of SARS-CoV-2 vaccines on PubMed, Web of Science, Cochrane Library, Google Scholar, Embase, Scopus, bioRxiv, and medRxiv published from November 26th, 2021, to June 27th, 2022 (full doses and the first booster), and to January 8th, 2023 (the second booster). The pooled VE against Omicron-associated infection and severe events were estimated. Results From 2,552 citations identified, 42 articles were included. The first booster provided stronger protection against Omicron than full doses alone, shown by VE estimates of 53.1% (95% CI: 48.0-57.8) vs. 28.6% (95% CI: 18.5-37.4) against infection and 82.5% (95% CI: 77.8-86.2) vs. 57.3% (95% CI: 48.5-64.7) against severe events. The second booster offered strong protection among adults within 60 days of vaccination against infection (VE=53.1%, 95% CI: 48.0-57.8) and severe events (VE=87.3% (95% CI: 75.5-93.4), comparable to the first booster with corresponding VE estimates of 59.9% against infection and 84.8% against severe events. The VE estimates of booster doses against severe events among adults sustained beyond 60 days, 77.6% (95% CI: 69.4-83.6) for first and 85.9% (95% CI: 80.3-89.9) for the second booster. The VE estimates against infection were less sustainable regardless of dose type. Pure mRNA vaccines provided comparable protection to partial mRNA vaccines, but both provided higher protection than non-mRNA vaccines. Conclusions One or two SARS-CoV-2 booster doses provide considerable protection against Omicron infection and substantial and sustainable protection against Omicron-induced severe clinical outcomes.
Collapse
Affiliation(s)
- Shangchen Song
- Department of Biostatistics, College of Public Health and Health professions and Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Zachary J. Madewell
- Department of Biostatistics, College of Public Health and Health professions and Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Mingjin Liu
- Department of Biostatistics, College of Public Health and Health professions and Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Ira M. Longini
- Department of Biostatistics, College of Public Health and Health professions and Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Yang Yang
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
14
|
Cortese P, Amato F, Davino A, De Franchis R, Esposito S, Zollo I, Di Domenico M, Solito E, Zarrilli F, Gentile L, Cernera G, Castaldo G. The Immune Response to SARS-CoV-2 Vaccine in a Cohort of Family Pediatricians from Southern Italy. Cells 2023; 12:1447. [PMID: 37296568 PMCID: PMC10252549 DOI: 10.3390/cells12111447] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
In Italy, from January 2021, the Ministry of Health indicated a vaccination plan against COVID for frail patients and physicians based on a three-dose scheme. However, conflicting results have been reported on which biomarkers permit immunization assessment. We used several laboratory approaches (i.e., antibodies serum levels, flow cytometry analysis, and cytokines release by stimulated cells) to investigate the immune response in a cohort of 53 family pediatricians (FPs) at different times after the vaccine. We observed that the BNT162b2-mRNA vaccine induced a significant increase of specific antibodies after the third (booster) dose; however, the antibody titer was not predictive of the risk of developing the infection in the six months following the booster dose. The antigen stimulation of PBMC cells from subjects vaccinated with the third booster jab induced the increase of the activated T cells (i.e., CD4+ CD154+); the frequency of CD4+ CD154+ TNF-α+ cells, as well as the TNF-α secretion, was not modified, while we observed a trend of increase of IFN-γ secretion. Interestingly, the level of CD8+ IFN-γ+ (independently from antibody titer) was significantly increased after the third dose and predicts the risk of developing the infection in the six months following the booster jab. Such results may impact also other virus vaccinations.
Collapse
Affiliation(s)
- Paolo Cortese
- Federazione Italiana Medici Pediatri (FIMP), 80142 Naples, Italy; (P.C.); (A.D.); (R.D.F.)
| | - Felice Amato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Antonio Davino
- Federazione Italiana Medici Pediatri (FIMP), 80142 Naples, Italy; (P.C.); (A.D.); (R.D.F.)
| | - Raffaella De Franchis
- Federazione Italiana Medici Pediatri (FIMP), 80142 Naples, Italy; (P.C.); (A.D.); (R.D.F.)
| | - Speranza Esposito
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Immacolata Zollo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Marina Di Domenico
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Egle Solito
- Centre for Translational Medicine and Therapeutics William Harvey Research Institute, Queen Mary Univesity, London E1 4NS, UK;
| | - Federica Zarrilli
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Laura Gentile
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Gustavo Cernera
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| | - Giuseppe Castaldo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy; (F.A.); (I.Z.); (F.Z.); (G.C.)
- CEINGE Biotecnologie Avanzate Franco Salvatore, Scarl, 80131 Naples, Italy; (S.E.); (L.G.)
| |
Collapse
|
15
|
Bobrovitz N, Ware H, Ma X, Li Z, Hosseini R, Cao C, Selemon A, Whelan M, Premji Z, Issa H, Cheng B, Abu Raddad LJ, Buckeridge DL, Van Kerkhove MD, Piechotta V, Higdon MM, Wilder-Smith A, Bergeri I, Feikin DR, Arora RK, Patel MK, Subissi L. Protective effectiveness of previous SARS-CoV-2 infection and hybrid immunity against the omicron variant and severe disease: a systematic review and meta-regression. THE LANCET. INFECTIOUS DISEASES 2023; 23:556-567. [PMID: 36681084 PMCID: PMC10014083 DOI: 10.1016/s1473-3099(22)00801-5] [Citation(s) in RCA: 251] [Impact Index Per Article: 251.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/01/2022] [Accepted: 11/21/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND The global surge in the omicron (B.1.1.529) variant has resulted in many individuals with hybrid immunity (immunity developed through a combination of SARS-CoV-2 infection and vaccination). We aimed to systematically review the magnitude and duration of the protective effectiveness of previous SARS-CoV-2 infection and hybrid immunity against infection and severe disease caused by the omicron variant. METHODS For this systematic review and meta-regression, we searched for cohort, cross-sectional, and case-control studies in MEDLINE, Embase, Web of Science, ClinicalTrials.gov, the Cochrane Central Register of Controlled Trials, the WHO COVID-19 database, and Europe PubMed Central from Jan 1, 2020, to June 1, 2022, using keywords related to SARS-CoV-2, reinfection, protective effectiveness, previous infection, presence of antibodies, and hybrid immunity. The main outcomes were the protective effectiveness against reinfection and against hospital admission or severe disease of hybrid immunity, hybrid immunity relative to previous infection alone, hybrid immunity relative to previous vaccination alone, and hybrid immunity relative to hybrid immunity with fewer vaccine doses. Risk of bias was assessed with the Risk of Bias In Non-Randomized Studies of Interventions Tool. We used log-odds random-effects meta-regression to estimate the magnitude of protection at 1-month intervals. This study was registered with PROSPERO (CRD42022318605). FINDINGS 11 studies reporting the protective effectiveness of previous SARS-CoV-2 infection and 15 studies reporting the protective effectiveness of hybrid immunity were included. For previous infection, there were 97 estimates (27 with a moderate risk of bias and 70 with a serious risk of bias). The effectiveness of previous infection against hospital admission or severe disease was 74·6% (95% CI 63·1-83·5) at 12 months. The effectiveness of previous infection against reinfection waned to 24·7% (95% CI 16·4-35·5) at 12 months. For hybrid immunity, there were 153 estimates (78 with a moderate risk of bias and 75 with a serious risk of bias). The effectiveness of hybrid immunity against hospital admission or severe disease was 97·4% (95% CI 91·4-99·2) at 12 months with primary series vaccination and 95·3% (81·9-98·9) at 6 months with the first booster vaccination after the most recent infection or vaccination. Against reinfection, the effectiveness of hybrid immunity following primary series vaccination waned to 41·8% (95% CI 31·5-52·8) at 12 months, while the effectiveness of hybrid immunity following first booster vaccination waned to 46·5% (36·0-57·3) at 6 months. INTERPRETATION All estimates of protection waned within months against reinfection but remained high and sustained for hospital admission or severe disease. Individuals with hybrid immunity had the highest magnitude and durability of protection, and as a result might be able to extend the period before booster vaccinations are needed compared to individuals who have never been infected. FUNDING WHO COVID-19 Solidarity Response Fund and the Coalition for Epidemic Preparedness Innovations.
Collapse
Affiliation(s)
- Niklas Bobrovitz
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Department of Critical Care Medicine, University of Calgary, Calgary, AB, Canada; Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Harriet Ware
- Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Xiaomeng Ma
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Zihan Li
- Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Bioengineering, University of California, Berkeley, CA, USA
| | - Reza Hosseini
- Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Christian Cao
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anabel Selemon
- Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mairead Whelan
- Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Zahra Premji
- Libraries, University of Victoria, Victoria, BC, Canada
| | - Hanane Issa
- Institute of Health Informatics, University College London, London, UK
| | - Brianna Cheng
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Laith J Abu Raddad
- Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Cornell University, Doha, Qatar
| | - David L Buckeridge
- Department of Epidemiology and Biostatistics, School of Population and Global Health, McGill University, Montreal, QC, Canada
| | | | - Vanessa Piechotta
- Department of Infectious Disease Epidemiology, Robert Koch Institute, Berlin, Germany
| | - Melissa M Higdon
- International Vaccine Access Center, Department of International Health, John Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Annelies Wilder-Smith
- Department of Immunizations, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland; Heidelberg Institute of Global Health, University of Heidelberg, Germany
| | - Isabel Bergeri
- Health Emergencies Programme, World Health Organization, Geneva, Switzerland
| | - Daniel R Feikin
- Department of Immunizations, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Rahul K Arora
- Centre for Health Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Minal K Patel
- Department of Immunizations, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Lorenzo Subissi
- Health Emergencies Programme, World Health Organization, Geneva, Switzerland
| |
Collapse
|
16
|
Boaventura VS, Cerqueira-Silva T, Barral-Netto M. The benefit of vaccination after previous SARS-CoV-2 infection in the omicron era. THE LANCET. INFECTIOUS DISEASES 2023; 23:511-512. [PMID: 36681085 PMCID: PMC9848411 DOI: 10.1016/s1473-3099(22)00880-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 01/20/2023]
Affiliation(s)
- Viviane S Boaventura
- LIB and LEITV Laboratories, Instituto Gonçalo Moniz, Fiocruz, Salvador, Bahia, Brazil,Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Thiago Cerqueira-Silva
- LIB and LEITV Laboratories, Instituto Gonçalo Moniz, Fiocruz, Salvador, Bahia, Brazil,Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Manoel Barral-Netto
- LIB and LEITV Laboratories, Instituto Gonçalo Moniz, Fiocruz, Salvador, Bahia, Brazil,Universidade Federal da Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
17
|
SARS-CoV-2 Seroprevalence among Canadian Blood Donors: The Advance of Omicron. Viruses 2022; 14:v14112336. [PMID: 36366432 PMCID: PMC9695729 DOI: 10.3390/v14112336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 02/01/2023] Open
Abstract
With the emergence of the SARS-CoV-2 Omicron variant in late 2021, Canadian public health case/contact testing was scaled back due to high infection rates with milder symptoms in a highly vaccinated population. We monitored the seroprevalence of SARS-CoV-2 nucleocapsid (anti-N) and spike protein (anti-S) antibodies in blood donors across Canada from September 2021 to June 2022 in 202,123 randomly selected samples. Multivariable logistic regression of anti-N positivity with month, age, sex, racialization, region, material and social deprivation (based on postal code) identified as independent predictors. Piece-wise logistic regression analysed the association between anti-S concentration and month, and anti-N/anti-S positivity. Infection-related seroprevalence (anti-N positive) was 4.38% (95% CI: 3.96, 4.81) in September reaching 50.70% (50.15, 52.16) in June; nearly 100% were anti-S positive throughout. Anti-N positivity was associated with younger age, male sex, the Alberta and Prairies regions, greater material deprivation and less social deprivation (p < 0.001). Anti-S concentration was high initially (3306 U/mL, IQR 4280 U/mL), increased to (13,659 U/mL, IQR 28,224 U/mL) by June (p < 0.001), following the pattern of deployment of the third and fourth vaccine doses and was higher in those that were anti-N positive (p < 0.001). Despite already high vaccination-related seroprevalence, infection-related seroprevalence increased dramatically with the emergence of the Omicron SARS-CoV-2 variant.
Collapse
|