1
|
Quach HQ, Haralambieva IH, Goergen KM, Grill DE, Chen J, Ovsyannikova IG, Poland GA, Kennedy RB. Similar humoral responses but distinct CD4 + T cell transcriptomic profiles in older adults elicited by MF59 adjuvanted and high dose influenza vaccines. Sci Rep 2024; 14:24420. [PMID: 39424894 PMCID: PMC11489691 DOI: 10.1038/s41598-024-75250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
Older age (≥ 65 years) is associated with impaired responses to influenza vaccination, leading to the preferential recommendation of MF59-adjuvanted (MF59Flu) or high-dose (HDFlu) influenza vaccines for this age group in the United States. Herein, we characterized transcriptomic profiles of CD4+ T cells isolated from 234 recipients (≥ 65 years) of either MF59Flu or HDFlu vaccine, prior to vaccination and 28 days thereafter. We identified 412 and 645 differentially expressed genes (DEGs) in CD4+ T cells of older adults after receiving MF59Flu and HDFlu, respectively. DEGs in CD4+ T cells of MF59Flu recipients were enriched in 14 KEGG pathways, all of which were downregulated. DEGs in CD4+ T cells of HDFlu recipients were enriched in 11 upregulated pathways and 20 downregulated pathways. CD4+ T cells in both vaccine groups shared 50 upregulated genes and 75 downregulated genes, all of which were enriched in 7 KEGG pathways. The remaining 287 and 520 DEGs were specifically associated with MF59Flu and HDFlu, respectively. Unexpectedly, none of these DEGs was significantly correlated with influenza A/H3N2-specific HAI titers, suggesting these DEGs at the individual level may have a limited role in protection against influenza. Our findings emphasize the need for further investigation into other factors influencing immunity against influenza in older adults.
Collapse
Affiliation(s)
- Huy Quang Quach
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Iana H Haralambieva
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Krista M Goergen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Diane E Grill
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Inna G Ovsyannikova
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Gregory A Poland
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA
| | - Richard B Kennedy
- Department of Internal Medicine, Vaccine Research Group, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
2
|
van der Heiden M, Shetty S, Bijvank E, Beckers L, Cevirgel A, van Sleen Y, Tcherniaeva I, Ollinger T, Burny W, van Binnendijk RS, van Houten MA, Buisman AM, Rots NY, van Beek J, van Baarle D. Multiple vaccine comparison in the same adults reveals vaccine-specific and age-related humoral response patterns: an open phase IV trial. Nat Commun 2024; 15:6603. [PMID: 39097574 PMCID: PMC11297912 DOI: 10.1038/s41467-024-50760-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/18/2024] [Indexed: 08/05/2024] Open
Abstract
Vaccine responsiveness is often reduced in older adults. Yet, our lack of understanding of low vaccine responsiveness hampers the development of effective vaccination strategies to reduce the impact of infectious diseases in the ageing population. Young-adult (25-49 y), middle-aged (50-64 y) and older-adult ( ≥ 65 y) participants of the VITAL clinical trials (n = 315, age-range: 28-98 y), were vaccinated with an annual (2019-2020) quadrivalent influenza (QIV) booster vaccine, followed by a primary 13-valent pneumococcal-conjugate (PCV13) vaccine (summer/autumn 2020) and a primary series of two SARS-CoV-2 mRNA-1273 vaccines (spring 2021). This unique setup allowed investigation of humoral responsiveness towards multiple vaccines within the same individuals over the adult age-range. Booster QIV vaccination induced comparable H3N2 hemagglutination inhibition (HI) titers in all age groups, whereas primary PCV13 and mRNA-1273 vaccination induced lower antibody concentrations in older as compared to younger adults (primary endpoint). The persistence of humoral responses, towards the 6 months timepoint, was shorter in older adults for all vaccines (secondary endpoint). Interestingly, highly variable vaccine responder profiles overarching multiple vaccines were observed. Yet, approximately 10% of participants, mainly comprising of older male adults, were classified as low responders to multiple vaccines. This study aids the identification of risk groups for low vaccine responsiveness and hence supports targeted vaccination strategies. Trial number: NL69701.041.19, EudraCT: 2019-000836-24.
Collapse
Affiliation(s)
- Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Sudarshan Shetty
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Elske Bijvank
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Lisa Beckers
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Alper Cevirgel
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Irina Tcherniaeva
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | | | - Rob S van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Marianne A van Houten
- Spaarne Academy, Spaarne Gasthuis, Hoofddorp, The Netherlands
- Department of Pediatrics, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Anne-Marie Buisman
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Nynke Y Rots
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
3
|
Cevirgel A, Shetty SA, Vos M, Nanlohy NM, Beckers L, Bijvank E, Rots N, van Beek J, Buisman A, van Baarle D. Pre-vaccination immunotypes reveal weak and robust antibody responders to influenza vaccination. Aging Cell 2024; 23:e14048. [PMID: 38146131 PMCID: PMC10861208 DOI: 10.1111/acel.14048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 12/27/2023] Open
Abstract
Effective vaccine-induced immune responses are particularly essential in older adults who face an increased risk of immunosenescence. However, the complexity and variability of the human immune system make predicting vaccine responsiveness challenging. To address this knowledge gap, our study aimed to characterize immune profiles that are predictive of vaccine responsiveness using "immunotypes" as an innovative approach. We analyzed an extensive set of innate and adaptive immune cell subsets in the whole blood of 307 individuals (aged 25-92) pre- and post-influenza vaccination which we associated with day 28 hemagglutination inhibition (HI) antibody titers. Building on our previous work that stratified individuals into nine immunotypes based on immune cell subsets, we identified two pre-vaccination immunotypes associated with weak and one showing robust day 28 antibody response. Notably, the weak responders demonstrated HLA-DR+ T-cell signatures, while the robust responders displayed a high naïve-to-memory T-cell ratio and percentage of nonclassical monocytes. These specific signatures deepen our understanding of the relationship between the baseline of the immune system and its functional potential. This approach could enhance our ability to identify individuals at risk of immunosenescence. Our findings highlight the potential of pre-vaccination immunotypes as an innovative tool for informing personalized vaccination strategies and improving health outcomes, particularly for aging populations.
Collapse
Affiliation(s)
- Alper Cevirgel
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
- Department of Medical Microbiology and Infection Prevention, Virology and Immunology research groupUniversity Medical Center GroningenGroningenThe Netherlands
| | - Sudarshan A. Shetty
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
- Department of Medical Microbiology and Infection Prevention, Virology and Immunology research groupUniversity Medical Center GroningenGroningenThe Netherlands
| | - Martijn Vos
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Nening M. Nanlohy
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Lisa Beckers
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Elske Bijvank
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Nynke Rots
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Josine van Beek
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Anne‐Marie Buisman
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Debbie van Baarle
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
- Department of Medical Microbiology and Infection Prevention, Virology and Immunology research groupUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
4
|
Bolton KJ, McCaw JM, Dafilis MP, McVernon J, Heffernan JM. Seasonality as a driver of pH1N12009 influenza vaccination campaign impact. Epidemics 2023; 45:100730. [PMID: 38056164 DOI: 10.1016/j.epidem.2023.100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/18/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023] Open
Abstract
Although the most recent respiratory virus pandemic was triggered by a Coronavirus, sustained and elevated prevalence of highly pathogenic avian influenza viruses able to infect mammalian hosts highlight the continued threat of pandemics of influenza A virus (IAV) to global health. Retrospective analysis of pandemic outcomes, including comparative investigation of intervention efficacy in different regions, provide important contributions to the evidence base for future pandemic planning. The swine-origin IAV pandemic of 2009 exhibited regional variation in onset, infection dynamics and annual infection attack rates (IARs). For example, the UK experienced three severe peaks of infection over two influenza seasons, whilst Australia experienced a single severe wave. We adopt a seasonally forced 2-subtype model for the transmission of pH1N12009 and seasonal H3N2 to examine the role vaccination campaigns may play in explaining differences in pandemic trajectories in temperate regions. Our model differentiates between the nature of vaccine- and infection-acquired immunity. In particular, we assume that immunity triggered by infection elicits heterologous cross-protection against viral shedding in addition to long-lasting neutralising antibody, whereas vaccination induces imperfect reduction in susceptibility. We employ an Approximate Bayesian Computation (ABC) framework to calibrate the model using data for pH1N12009 seroprevalence, relative subtype dominance, and annual IARs for Australia and the UK. Heterologous cross-protection substantially suppressed the pandemic IAR over the posterior, with the strength of protection against onward transmission inversely correlated with the initial reproduction number. We show that IAV pandemic timing relative to the usual seasonal influenza cycle influenced the size of the initial waves of pH1N12009 in temperate regions and the impact of vaccination campaigns.
Collapse
Affiliation(s)
- Kirsty J Bolton
- School of Mathematical Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| | - James M McCaw
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Mathew P Dafilis
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Jodie McVernon
- Peter Doherty Institute for Infection and Immunity, The Royal Melbourne Hospital and The University of Melbourne, Parkville, Australia
| | - Jane M Heffernan
- Centre for Disease Modelling, Mathematics & Statistics, York University, Canada
| |
Collapse
|
5
|
Lewis ED, Crowley DC, Guthrie N, Evans M. Role of Acacia catechu and Scutellaria baicalensis in Enhancing Immune Function Following Influenza Vaccination of Healthy Adults: A Randomized, Triple-Blind, Placebo-Controlled Clinical Trial. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2023; 42:678-690. [PMID: 36413261 DOI: 10.1080/27697061.2022.2145525] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE The study aimed to examine the role of an Acacia catechu and Scutellaria baicalensis formulation, UP446, on supporting immune function in response to influenza vaccination. METHODS A randomized, triple-blind, placebo-controlled, parallel study consisted of a 56-day intervention period with a 28-day pre-vaccination period, an influenza vaccination on Day 28 and 28-day post-vaccination period. Fifty healthy adults 40-80 years of age who had not received their flu vaccine were randomized to either UP446 or Placebo. At baseline, Days 28 and 56, immune and oxidative stress markers were measured in blood and a quality of life questionnaire was administered. Participants completed the Wisconsin Upper Respiratory Symptom Survey (WURSS)-24 daily. RESULTS In the post-vaccination period, total IgA and IgG levels increased in participants supplemented with UP446 vs. those on Placebo (p ≤ 0.026). As well, influenza B-specific IgG increased 19.4% from Day 28 to 56 and 11.6% from baseline at Day 56 (p ≤ 0.0075). Serum glutathione peroxidase (GSH-Px) was increased in the pre-vaccination period and from baseline at Day 56 with UP446 supplementation (p ≤ 0.0270). CONCLUSION These results suggest a 56-day supplementation with UP446 was beneficial in mounting a robust humoral response following vaccination. Increasing GSH-Px in the pre-vaccination period may help improve antioxidant functions and potentially mitigate the oxidative stress induced following vaccination.
Collapse
|
6
|
Dias Assis BR, Gomes IP, de Castro JT, Rivelli GG, de Castro NS, Gomez-Mendoza DP, Bagno FF, Hojo-Souza NS, Chaves Maia AL, Lages EB, da Fonseca FG, Ribeiro Teixeira SM, Fernandes AP, Gazzinelli RT, Castro Goulart GA. Quality attributes of CTVad1, a nanoemulsified adjuvant for phase I clinical trial of SpiN COVID-19 vaccine. Nanomedicine (Lond) 2023; 18:1175-1194. [PMID: 37712604 DOI: 10.2217/nnm-2023-0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Aim: To develop, characterize and evaluate an oil/water nanoemulsion with squalene (CTVad1) to be approved as an adjuvant for the SpiN COVID-19 vaccine clinical trials. Materials & methods: Critical process parameters (CPPs) of CTVad1 were standardized to meet the critical quality attributes (CQAs) of an adjuvant for human use. CTVad1 and the SpiN-CTVad1 vaccine were submitted to physicochemical, stability, in vitro and in vivo studies. Results & conclusion: All CQAs were met in the CTVad1 production process. SpiN- CTVad1 met CQAs and induced high levels of antibodies and specific cellular responses in in vivo studies. These results represented a critical step in the process developed to meet regulatory requirements for the SpiN COVID-19 vaccine clinical trial.
Collapse
Affiliation(s)
- Bruna Rodrigues Dias Assis
- Department of Pharmaceuticals, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Isabela Pereira Gomes
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Júlia Teixeira de Castro
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Graziella Gomes Rivelli
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Natália Salazar de Castro
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Diana Paola Gomez-Mendoza
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Flávia Fonseca Bagno
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Natália Satchiko Hojo-Souza
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
| | - Ana Luiza Chaves Maia
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Eduardo Burgarelli Lages
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| | - Flávio Guimaraes da Fonseca
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Santuza Maria Ribeiro Teixeira
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana Paula Fernandes
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Department of Clinical & Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ricardo Tostes Gazzinelli
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz-Minas, Belo Horizonte, MG, 30190-002, Brazil
- Department of Biochemistry & Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Gisele Assis Castro Goulart
- Department of Pharmaceuticals, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
- Centro de Tecnologia de Vacinas da Universidade Federal de Minas Gerais, Belo Horizonte, Belo Horizonte, MG, 31310-260, Brazil
| |
Collapse
|
7
|
Gianchecchi E, Torelli A, Piu P, Bonifazi C, Ganfini L, Montomoli E. Flow cytometry as an integrative method for the evaluation of vaccine immunogenicity: A validation approach. Biochem Biophys Rep 2023; 34:101472. [PMID: 37153861 PMCID: PMC10160688 DOI: 10.1016/j.bbrep.2023.101472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023] Open
Abstract
The applied bioanalytical assays used for the evaluation of human immune responses from samples collected during clinical trials must be well characterized, fully validated and properly documented to provide reliable results. Even though recommendations for the standardization of flow cytometry instrumentation and assay validation for its clinical application have been published by several organizations, definitive guidelines are not available yet. The aim of the present paper is to provide a validation approach for flow cytometry, examining parameters such as linearity, relative accuracy, repeatability, intermediate precision, range and detection limits and specificity, in order to demonstrate and document its applicability for clinical research purposes and its possible use as one of the methods for the evaluation of vaccine immunogenicity.
Collapse
Affiliation(s)
| | - Alessandro Torelli
- CSL Seqirus, Via Del Pozzo 3/A, S. Martino, 53035, Monteriggioni, SI, Italy
| | | | - Carolina Bonifazi
- VisMederi S.r.l., Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Emanuele Montomoli
- VisMederi S.r.l., Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
8
|
Distinct immunological and molecular signatures underpinning influenza vaccine responsiveness in the elderly. Nat Commun 2022; 13:6894. [PMID: 36371426 PMCID: PMC9653450 DOI: 10.1038/s41467-022-34487-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Seasonal influenza outbreaks, especially in high-risk groups such as the elderly, represent an important public health problem. Prevailing inadequate efficacy of seasonal vaccines is a crucial bottleneck. Understanding the immunological and molecular mechanisms underpinning differential influenza vaccine responsiveness is essential to improve vaccination strategies. Here we show comprehensive characterization of the immune response of randomly selected elderly participants (≥ 65 years), immunized with the adjuvanted influenza vaccine Fluad. In-depth analyses by serology, multi-parametric flow cytometry, multiplex and transcriptome analysis, coupled to bioinformatics and mathematical modelling, reveal distinguishing immunological and molecular features between responders and non-responders defined by vaccine-induced seroconversion. Non-responders are specifically characterized by multiple suppressive immune mechanisms. The generated comprehensive high dimensional dataset enables the identification of putative mechanisms and nodes responsible for vaccine non-responsiveness independently of confounding age-related effects, with the potential to facilitate development of tailored vaccination strategies for the elderly.
Collapse
|
9
|
Pieren DKJ, Smits NAM, Postel RJ, Kandiah V, de Wit J, van Beek J, van Baarle D, Guichelaar T. Co-Expression of TIGIT and Helios Marks Immunosenescent CD8+ T Cells During Aging. Front Immunol 2022; 13:833531. [PMID: 35651622 PMCID: PMC9148977 DOI: 10.3389/fimmu.2022.833531] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aging leads to alterations in the immune system that result in ineffective responsiveness against pathogens. Features of this process, collectively known as immunosenescence, accumulate in CD8+ T cells with age and have been ascribed to differentiation of these cells during the course of life. Here we aimed to identify novel markers in CD8+ T cells associated with immunosenescence. Furthermore, we assessed how these markers relate to the aging-related accumulation of highly differentiated CD27-CD28- cells. We found that co-expression of the transcription factor Helios and the aging-related marker TIGIT identifies CD8+ T cells that fail to proliferate and show impaired induction of activation markers CD69 and CD25 in response to stimulation in vitro. Despite this, in blood of older adults we found TIGIT+Helios+ T cells to become highly activated during an influenza-A virus infection, but these higher frequencies of activated TIGIT+Helios+ T cells associate with longer duration of coughing. Moreover, in healthy individuals, we found that TIGIT+Helios+ CD8+ T cells accumulate with age in the highly differentiated CD27-CD28- population. Interestingly, TIGIT+Helios+ CD8+ T cells also accumulate with age among the less differentiated CD27+CD28- T cells before their transit into the highly differentiated CD27-CD28- stage. This finding suggests that T cells with immunosenescent features become prominent at old age also within the earlier differentiation states of these cells. Our findings show that co-expression of TIGIT and Helios refines the definition of immunosenescent CD8+ T cells and challenge the current dogma of late differentiation stage as proxy for T-cell immunosenescence.
Collapse
Affiliation(s)
- Daan K. J. Pieren
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Noortje A. M. Smits
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Rimke J. Postel
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Vinitha Kandiah
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
| | - Debbie van Baarle
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Teun Guichelaar
- Centre for Infectious Disease Control, National Institute for Public Health and The Environment, Bilthoven, Netherlands
- *Correspondence: Teun Guichelaar,
| |
Collapse
|
10
|
Kaaijk P, Olivo Pimentel V, Emmelot ME, Poelen MCM, Cevirgel A, Schepp RM, den Hartog G, Reukers DF, Beckers L, van Beek J, van Els CACM, Meijer A, Rots NY, de Wit J. Children and Adults With Mild COVID-19: Dynamics of the Memory T Cell Response up to 10 Months. Front Immunol 2022; 13:817876. [PMID: 35197982 PMCID: PMC8858984 DOI: 10.3389/fimmu.2022.817876] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/18/2022] [Indexed: 12/14/2022] Open
Abstract
BackgroundSevere acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has led to considerable morbidity/mortality worldwide, but most infections, especially among children, have a mild course. However, it remains largely unknown whether infected children develop cellular immune memory.MethodsTo determine whether a memory T cell response is being developed, we performed a longitudinal assessment of the SARS-CoV-2-specific T cell response by IFN-γ ELISPOT and activation marker analyses of peripheral blood samples from unvaccinated children and adults with mild-to-moderate COVID-19.ResultsUpon stimulation of PBMCs with heat-inactivated SARS-CoV-2 or overlapping peptides of spike (S-SARS-CoV-2) and nucleocapsid proteins, we found S-SARS-CoV-2-specific IFN-γ T cell responses in infected children (83%) and adults (100%) that were absent in unexposed controls. Frequencies of SARS-CoV-2-specific T cells were higher in infected adults, especially in those with moderate symptoms, compared to infected children. The S-SARS-CoV-2 IFN-γ T cell response correlated with S1-SARS-CoV-2-specific serum antibody concentrations. Predominantly, effector memory CD4+ T cells of a Th1 phenotype were activated upon exposure to SARS-CoV-2 antigens. Frequencies of SARS-CoV-2-specific T cells were significantly reduced at 10 months after symptom onset, while S1-SARS-CoV-2-specific IgG concentrations were still detectable in 90% of all children and adults.ConclusionsOur data indicate that an antigen-specific T cell and antibody response is developed after mild SARS-CoV-2 infection in children and adults. It remains to be elucidated to what extent this SARS-CoV-2-specific response can contribute to an effective recall response after reinfection.
Collapse
Affiliation(s)
- Patricia Kaaijk
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- *Correspondence: Patricia Kaaijk,
| | - Verónica Olivo Pimentel
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Maarten E. Emmelot
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Martien C. M. Poelen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Alper Cevirgel
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Rutger M. Schepp
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Gerco den Hartog
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Daphne F.M. Reukers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Lisa Beckers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Cécile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Adam Meijer
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Nynke Y. Rots
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
11
|
Zorgi NE, Meireles LR, Oliveira DBL, Araujo DB, Durigon EL, Andrade Junior HFD. Isolated specific IgA against respiratory viruses, Influenza or SARS-CoV-2, present in the saliva of a fraction of healthy and asymptomatic volunteers. Clinics (Sao Paulo) 2022; 77:100105. [PMID: 36116267 PMCID: PMC9444893 DOI: 10.1016/j.clinsp.2022.100105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Defense against respiratory viruses depends on an immune response present in the mucosa, as saliva IgA secretes antibodies. During the pandemic, such as influenza or SARS-CoV-2, most infected patients are asymptomatic but retain specific antibodies post-infection. The authors evaluated IgG and IgA antibodies against SARS-CoV-2 and influenza in the saliva of asymptomatic volunteers, validated with controls or vaccinated individuals. METHODS The authors detected specific antibodies by validated conventional ELISA using natural SARS-CoV-2 antigens from infected Vero cells or capture-ELISA for influenza using natural antigens of the influenza vaccine. RESULTS Saliva from influenza-vaccinated individuals had more IgA than paired serum, contrary to the findings for specific IgG. In COVID-19-vaccinated samples, specific IgA in saliva increased after vaccination, but IgG levels were high after the first dose. In saliva from the asymptomatic population (226), anti-Influenza IgG was found in 57.5% (130) of samples, higher than IgA, found in 35% (79) of samples. IgA results were similar for SARS-CoV-2, with IgA present in 30% (68) of samples, while IgG was less present, in 44.2% (100) of samples. The proportion of influenza IgG responders was higher than that for SARS-CoV-2 IgG, but both populations presented similar proportions of IgA responders, possibly due to variable memory B cell survival. For both viruses, the authors found an important proportion (> 10%) of IgA+IgG- samples, suggesting the occurrence of humoral immunity directed to the mucosa. CONCLUSION Specific antibodies for respiratory viruses in saliva are found in either infection or vaccination and are a convenient and sensitive diagnostic tool for host immune response.
Collapse
Affiliation(s)
- Nahiara Esteves Zorgi
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil
| | - Luciana R Meireles
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil
| | | | - Danielle Bastos Araujo
- Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Edson L Durigon
- Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Heitor Franco de Andrade Junior
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil.
| |
Collapse
|
12
|
Pieren DKJ, Smits NAM, Hoeboer J, Kandiah V, Postel RJ, Mariman R, Beek J, Baarle D, Wit J, Guichelaar T. Regulatory KIR + RA + T cells accumulate with age and are highly activated during viral respiratory disease. Aging Cell 2021; 20:e13372. [PMID: 34043881 PMCID: PMC8208794 DOI: 10.1111/acel.13372] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/18/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Severe respiratory viral infectious diseases such as influenza and COVID‐19 especially affect the older population. This is partly ascribed to diminished CD8+ T‐cell responses a result of aging. The phenotypical diversity of the CD8+ T‐cell population has made it difficult to identify the impact of aging on CD8+ T‐cell subsets associated with diminished CD8+ T‐cell responses. Here we identify a novel human CD8+ T‐cell subset characterized by expression of Killer‐cell Immunoglobulin‐like Receptors (KIR+) and CD45RA (RA+). These KIR+RA+ T cells accumulated with age in the blood of healthy individuals (20–82 years of age, n = 50), expressed high levels of aging‐related markers of T‐cell regulation, and were functionally capable of suppressing proliferation of other CD8+ T cells. Moreover, KIR+RA+ T cells were a major T‐cell subset becoming activated in older adults suffering from an acute respiratory viral infection (n = 36), including coronavirus and influenza virus infection. In addition, older adults with influenza A infection showed that higher activation status of their KIR+RA+ T cells associated with longer duration of respiratory symptoms. Together, our data indicate that KIR+RA+ T cells are a unique human T‐cell subset with regulatory properties that may explain susceptibility to viral respiratory disease at old age.
Collapse
Affiliation(s)
- Daan K. J. Pieren
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Noortje A. M. Smits
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Jeroen Hoeboer
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Vinitha Kandiah
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Rimke J. Postel
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Rob Mariman
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Josine Beek
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Debbie Baarle
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
- Center for Translational Immunology University Medical Center Utrecht Utrecht University Utrecht The Netherlands
| | - Jelle Wit
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| | - Teun Guichelaar
- Centre for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven The Netherlands
| |
Collapse
|
13
|
van den Berg SPH, Lanfermeijer J, Jacobi RHJ, Hendriks M, Vos M, van Schuijlenburg R, Nanlohy NM, Borghans JAM, van Beek J, van Baarle D, de Wit J. Latent CMV Infection Is Associated With Lower Influenza Virus-Specific Memory T-Cell Frequencies, but Not With an Impaired T-Cell Response to Acute Influenza Virus Infection. Front Immunol 2021; 12:663664. [PMID: 34025665 PMCID: PMC8131658 DOI: 10.3389/fimmu.2021.663664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Latent infection with cytomegalovirus (CMV) is assumed to contribute to the age-associated decline of the immune system. CMV induces large changes in the T-cell pool and may thereby affect other immune responses. CMV is expected to impact especially older adults, who are already at higher risk of severe disease and hospitalization upon infections such as influenza virus (IAV) infection. Here, we investigated the impact of CMV infection on IAV-specific CD8+ T-cell frequencies in healthy individuals (n=96) and the response to IAV infection in older adults (n=72). IAV-specific memory T-cell frequencies were lower in healthy CMV+ older individuals compared to healthy CMV- older individuals. Upon acute IAV infection, CMV serostatus or CMV-specific antibody levels were not negatively associated with IAV-specific T-cell frequencies, function, phenotype or T-cell receptor repertoire diversity. This suggests that specific T-cell responses upon acute IAV infection are not negatively affected by CMV. In addition, we found neither an association between CMV infection and inflammatory cytokine levels in serum during acute IAV infection nor between cytokine levels and the height of the IAV-specific T-cell response upon infection. Finally, CMV infection was not associated with increased severity of influenza-related symptoms. In fact, CMV infection was even associated with increased IAV-specific T-cell responses early upon acute IAV infection. In conclusion, although associated with lower frequencies of memory IAV-specific T cells in healthy individuals, CMV infection does not seem to hamper the induction of a proper T-cell response during acute IAV infection in older adults.
Collapse
Affiliation(s)
- Sara P H van den Berg
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Josien Lanfermeijer
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ronald H J Jacobi
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Marion Hendriks
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Martijn Vos
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Roos van Schuijlenburg
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Nening M Nanlohy
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - José A M Borghans
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Debbie van Baarle
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jelle de Wit
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| |
Collapse
|
14
|
Lanfermeijer J, de Greef PC, Hendriks M, Vos M, van Beek J, Borghans JAM, van Baarle D. Age and CMV-Infection Jointly Affect the EBV-Specific CD8 + T-Cell Repertoire. FRONTIERS IN AGING 2021; 2:665637. [PMID: 35822032 PMCID: PMC9261403 DOI: 10.3389/fragi.2021.665637] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/31/2021] [Indexed: 01/15/2023]
Abstract
CD8+ T cells play an important role in protection against viral infections. With age, changes in the T-cell pool occur, leading to diminished responses against both new and recurring infections in older adults. This is thought to be due to a decrease in both T-cell numbers and T-cell receptor (TCR) diversity. Latent infection with cytomegalovirus (CMV) is assumed to contribute to this age-associated decline of the immune system. The observation that the level of TCR diversity in the total memory T-cell pool stays relatively stable during aging is remarkable in light of the constant input of new antigen-specific memory T cells. What happens with the diversity of the individual antigen-specific T-cell repertoires in the memory pool remains largely unknown. Here we studied the effect of aging on the phenotype and repertoire diversity of CMV-specific and Epstein-Barr virus (EBV)-specific CD8+ T cells, as well as the separate effects of aging and CMV-infection on the EBV-specific T-cell repertoire. Antigen-specific T cells against both persistent viruses showed an age-related increase in the expression of markers associated with a more differentiated phenotype, including KLRG-1, an increase in the fraction of terminally differentiated T cells, and a decrease in the diversity of the T-cell repertoire. Not only age, but also CMV infection was associated with a decreased diversity of the EBV-specific T-cell repertoire. This suggests that both CMV infection and age can impact the T-cell repertoire against other antigens.
Collapse
Affiliation(s)
- Josien Lanfermeijer
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Peter C. de Greef
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | - Marion Hendriks
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Martijn Vos
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - José A. M. Borghans
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Debbie van Baarle
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
15
|
Immunogenicity of standard, high-dose, MF59-adjuvanted, and recombinant-HA seasonal influenza vaccination in older adults. NPJ Vaccines 2021; 6:25. [PMID: 33594050 PMCID: PMC7886864 DOI: 10.1038/s41541-021-00289-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/15/2021] [Indexed: 12/18/2022] Open
Abstract
The vaccine efficacy of standard-dose seasonal inactivated influenza vaccines (S-IIV) can be improved by the use of vaccines with higher antigen content or adjuvants. We conducted a randomized controlled trial in older adults to compare cellular and antibody responses of S-IIV versus enhanced vaccines (eIIV): MF59-adjuvanted (A-eIIV), high-dose (H-eIIV), and recombinant-hemagglutinin (HA) (R-eIIV). All vaccines induced comparable H3-HA-specific IgG and elevated antibody-dependent cellular cytotoxicity (ADCC) activity at day 30 post vaccination. H3-HA-specific ADCC responses were greatest following H-eIIV. Only A-eIIV increased H3-HA-IgG avidity, HA-stalk IgG and ADCC activity. eIIVs also increased polyfunctional CD4+ and CD8+ T cell responses, while cellular immune responses were skewed toward single-cytokine-producing T cells among S-IIV subjects. Our study provides further immunological evidence for the preferential use of eIIVs in older adults as each vaccine platform had an advantage over the standard-dose vaccine in terms of NK cell activation, HA-stalk antibodies, and T cell responses.
Collapse
|
16
|
Strizova Z, Smetanova J, Bartunkova J, Milota T. Principles and Challenges in anti-COVID-19 Vaccine Development. Int Arch Allergy Immunol 2021; 182:339-349. [PMID: 33524979 PMCID: PMC7900461 DOI: 10.1159/000514225] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/30/2020] [Indexed: 12/05/2022] Open
Abstract
The number of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients keeps rising in most of the European countries despite the pandemic precaution measures. The current antiviral and anti-inflammatory therapeutic approaches are only supportive, have limited efficacy, and the prevention in reducing the transmission of SARS-CoV-2 virus is the best hope for public health. It is presumed that an effective vaccination against SARS-CoV-2 infection could mobilize the innate and adaptive immune responses and provide a protection against severe forms of coronavirus disease 2019 (COVID-19) disease. As the race for the effective and safe vaccine has begun, different strategies were introduced. To date, viral vector-based vaccines, genetic vaccines, attenuated vaccines, and protein-based vaccines are the major vaccine types tested in the clinical trials. Over 80 clinical trials have been initiated; however, only 18 vaccines have reached the clinical phase II/III or III, and 4 vaccine candidates are under consideration or have been approved for the use so far. In addition, the protective effect of the off-target vaccines, such as Bacillus Calmette-Guérin and measles vaccine, is being explored in randomized prospective clinical trials with SARS-CoV-2-infected patients. In this review, we discuss the most promising anti-COVID-19 vaccine clinical trials and different vaccination strategies in order to provide more clarity into the ongoing clinical trials.
Collapse
Affiliation(s)
- Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jitka Smetanova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Jirina Bartunkova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomas Milota
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia,
- Department of Paediatric and Adult Rheumatology, University Hospital Motol, Prague, Czechia,
| |
Collapse
|
17
|
Di Blasi D, Claessen I, Turksma AW, van Beek J, Ten Brinke A. Guidelines for analysis of low-frequency antigen-specific T cell results: Dye-based proliferation assay vs 3H-thymidine incorporation. J Immunol Methods 2020; 487:112907. [PMID: 33152332 DOI: 10.1016/j.jim.2020.112907] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
It is generally recognized that dysregulation of the immune system plays a critical role in many diseases, including autoimmune diseases and cancer. T cells play a crucial role in maintaining self-tolerance, while loss of immune tolerance and T cell activation can lead to severe inflammation and tissue damage. T cell responses have a key role in the effectiveness of vaccination strategies and immunomodulating therapies. Immunomonitoring methods have the ability to elucidate immunological processes, monitor the development of disease and assess therapeutic effects. In this respect, it is of particular interest to evaluate antigen (Ag)-specific T cells by determining their frequency, type and functionality in cellular assays. Nevertheless, Ag-specific T cells are detected infrequently in most diseases using current techniques. Many efforts have been made to develop more sensitive, reproducible, and reliable methods for Ag-specific T cell detection. It has been found that analysis of cellular proliferation can be a useful tool to determine the presence and frequency of Ag-specific T cell and to provides insight into modulation of the T cell response by a specific antigen or therapy. However, the selection of a cut-off value for a positive response and therefore a more accurate interpretation of the data, continues to be a major concern. Here, we provide guidelines to select a proper cut-off for monitoring of Ag-specific CD4+ T cell responses. In vitro Ag-stimulation has been assessed with two methods; a dye-based proliferation assay and 3H-thymidine-based assay. Two cut-off approaches are compared; mean and variance of control wells, and the stimulation index. By evaluating the proliferative response to the in vitro Ag-stimulation using these two methods, we demonstrate the importance of taking into consideration the variability of the control wells to distinguish a positive from a false positive response.
Collapse
Affiliation(s)
- Daniela Di Blasi
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands.
| | - Iris Claessen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands; Sanquin Diagnostics B.V., Amsterdam, the Netherlands
| | | | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, the Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, AMC, Amsterdam, the Netherlands.
| |
Collapse
|
18
|
Elicitation of integrated immunity in mice by a novel pneumococcal polysaccharide vaccine conjugated with HBV surface antigen. Sci Rep 2020; 10:6470. [PMID: 32286332 PMCID: PMC7156719 DOI: 10.1038/s41598-020-62185-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/10/2020] [Indexed: 12/25/2022] Open
Abstract
The conjugation of polysaccharides with an effective carrier protein is critical for the development of effective bacterial polysaccharide vaccines. Therefore, the identification and optimization of carrier proteins to induce an effective immune response is necessary for developing a combined vaccine. In the current study, we utilized hepatitis B virus surface antigen (HBsAg) as a novel carrier protein combined with a capsular polysaccharide molecule to develop a new pneumococcal conjugated vaccine. The specific antibodies and T cell immune response against the capsular polysaccharide and HBsAg in the mice immunized with this conjugated vaccine were evaluated. In addition, the unique gene profiles of immune cells induced by this conjugated vaccine in the immunized mice were analyzed. Our results demonstrated that the vaccine consisting of pneumonia type 33 F capsular polysaccharide (Pn33Fps) conjugated with HBsAg can induce strong specific immune responses against both antigens in vivo in immunized mice. Furthermore, the conjugated vaccine induced higher expression of genes related to the activation of immunity and higher antibody titers against Pn33Fps and HBsAg in mice than those obtained via vaccination with a single antigen. Analyses of the dynamic expression changes in immunity-related genes in mice immunized with Pn33Fps_HBs, Pn33Fps, or HBsAg indicated the potent immunogenicity of the conjugated vaccine. In addition, a pathological evaluation of the organs from immunized mice further suggested that the conjugated vaccine is safe. Together, these results indicate that a conjugated vaccine consisting of Pn33Fps with HBsAg is a novel and effective vaccine.
Collapse
|
19
|
A Recombinant Influenza A/H1N1 Carrying A Short Immunogenic Peptide of MERS-CoV as Bivalent Vaccine in BALB/c Mice. Pathogens 2019; 8:pathogens8040281. [PMID: 31810359 PMCID: PMC6963271 DOI: 10.3390/pathogens8040281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/21/2019] [Accepted: 11/30/2019] [Indexed: 01/26/2023] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) became a global human health threat since its first documentation in humans in 2012. An efficient vaccine for the prophylaxis of humans in hotspots of the infection (e.g., Saudi Arabia) is necessary but no commercial vaccines are yet approved. In this study, a chimeric DNA construct was designed to encode an influenza A/H1N1 NA protein which is flanking immunogenic amino acids (aa) 736–761 of MERS-CoV spike protein. Using the generated chimeric construct, a novel recombinant vaccine strain against pandemic influenza A virus (H1N1pdm09) and MERS-CoV was generated (chimeric bivalent 5 + 3). The chimeric bivalent 5 + 3 vaccine strain comprises a recombinant PR8-based vaccine, expressing the PB1, HA, and chimeric NA of pandemic 2009 H1N1. Interestingly, an increase in replication efficiency of the generated vaccine strain was observed when compared to the PR8-based 5 + 3 H1N1pdm09 vaccine strain that lacks the MERS-CoV spike peptide insert. In BALB/c mice, the inactivated chimeric bivalent vaccine induced potent and specific neutralizing antibodies against MERS-CoV and H1N1pdm09. This novel approach succeeded in developing a recombinant influenza virus with potential use as a bivalent vaccine against H1N1pdm09 and MERS-CoV. This approach provides a basis for the future development of chimeric influenza-based vaccines against MERS-CoV and other viruses.
Collapse
|
20
|
Shah NM, Imami N, Kelleher P, Barclay WS, Johnson MR. Pregnancy-related immune suppression leads to altered influenza vaccine recall responses. Clin Immunol 2019; 208:108254. [PMID: 31470087 DOI: 10.1016/j.clim.2019.108254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/17/2019] [Accepted: 08/26/2019] [Indexed: 01/12/2023]
Abstract
Pregnancy is a risk factor for severe influenza infection. Despite achieving seroprotective antibody titres post immunisation fewer pregnant women experience a reduction in influenza-like illness compared to non-pregnant cohorts. This may be due to the effects that immune-modulation in pregnancy has on vaccine efficacy leading to a less favourable immunologic response. To understand this, we investigated the antigen-specific cellular responses and leukocyte phenotype in pregnant and non-pregnant women who achieved seroprotection post immunisation. We show that pregnancy is associated with better antigen-specific inflammatory (IFN-γ) responses and an expansion of central memory T cells (Tcm) post immunisation, but low-level pregnancy-related immune regulation (HLA-G, PIBF) and associated reduced B-cell antibody maintenance (TGF-β) suggest poor immunologic responses compared to the non-pregnant. Thus far, studies of influenza vaccine immunogenicity have focused on the induction of antibodies but understanding additional vaccine-related cellular responses is needed to fully appreciate how pregnancy impacts on vaccine effectiveness.
Collapse
Affiliation(s)
- Nishel M Shah
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom.
| | - Nesrina Imami
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Peter Kelleher
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Wendy S Barclay
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Mark R Johnson
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| |
Collapse
|