1
|
Amarin JZ, Dulek DE, Simmons J, Hayek H, Chappell JD, Nochowicz CH, Kitko CL, Schuster JE, Muñoz FM, Bocchini CE, Moulton EA, Coffin SE, Freedman JL, Ardura MI, Wattier RL, Maron G, Grimley M, Paulsen G, Danziger-Isakov L, Carpenter PA, Englund JA, Halasa NB, Spieker AJ, Kalams SA. Immunophenotypic predictors of influenza vaccine immunogenicity in pediatric hematopoietic cell transplant recipients. Blood Adv 2024; 8:1880-1892. [PMID: 38386973 PMCID: PMC11007439 DOI: 10.1182/bloodadvances.2023012118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
ABSTRACT Pediatric hematopoietic cell transplant (HCT) recipients exhibit poor serologic responses to influenza vaccination early after transplant. To facilitate the optimization of influenza vaccination timing, we sought to identify B- and T-cell subpopulations associated with influenza vaccine immunogenicity in this population. We used mass cytometry to phenotype peripheral blood mononuclear cells collected from pediatric HCT recipients enrolled in a multicenter influenza vaccine trial comparing high- and standard-dose formulations over 3 influenza seasons (2016-2019). We fit linear regression models to estimate relationships between immune cell subpopulation numbers before vaccination and prevaccination to postvaccination geometric mean fold rises in antigen-specific (A/H3N2, A/H1N1, and B/Victoria) serum hemagglutination inhibition antibody titers (28-42 days, and ∼6 months after 2 doses). For cell subpopulations identified as predictive of a response to all 3 antigens, we conducted a sensitivity analysis including time after transplant as an additional covariate. Among 156 HCT recipients, we identified 33 distinct immune cell subpopulations; 7 significantly predicted responses to all 3 antigens 28 to 42 days after a 2-dose vaccine series, irrespective of vaccine dose. We also found evidence that baseline absolute numbers of naïve B cells, naïve CD4+ T cells, and circulating T follicular helper cells predicted peak and sustained vaccine-induced titers irrespective of dose or timing of posttransplant vaccine administration. In conclusion, several B- and T-cell subpopulations predicted influenza vaccine immunogenicity in pediatric HCT recipients. This study provides insights into the immune determinants of vaccine responses and may help guide the development of tailored vaccination strategies for this vulnerable population.
Collapse
Affiliation(s)
- Justin Z. Amarin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
- Epidemiology Doctoral Program, School of Medicine, Vanderbilt University, Nashville, TN
| | - Daniel E. Dulek
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Joshua Simmons
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Haya Hayek
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - James D. Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | | | - Carrie L. Kitko
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | | | - Flor M. Muñoz
- Division of Infectious Diseases, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
- Department of Molecular Virology and Microbiology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
| | - Claire E. Bocchini
- Division of Infectious Diseases, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
| | - Elizabeth A. Moulton
- Division of Infectious Diseases, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
| | - Susan E. Coffin
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jason L. Freedman
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Monica I. Ardura
- Division of Infectious Diseases and Host Defense Program, Nationwide Children’s Hospital, Columbus, OH
- Department of Pediatrics, The Ohio State University, Columbus, OH
| | - Rachel L. Wattier
- Department of Pediatrics, University of California San Francisco and Benioff Children’s Hospital, San Francisco, CA
| | - Gabriela Maron
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN
| | - Michael Grimley
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Grant Paulsen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Lara Danziger-Isakov
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Paul A. Carpenter
- Department of Pediatrics, University of Washington and Seattle Children’s Research Institute, Seattle, WA
| | - Janet A. Englund
- Department of Pediatrics, University of Washington and Seattle Children’s Research Institute, Seattle, WA
| | - Natasha B. Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Andrew J. Spieker
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Spyros A. Kalams
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
| |
Collapse
|
2
|
Arunachalam AB. Vaccines Induce Homeostatic Immunity, Generating Several Secondary Benefits. Vaccines (Basel) 2024; 12:396. [PMID: 38675778 PMCID: PMC11053716 DOI: 10.3390/vaccines12040396] [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: 02/27/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The optimal immune response eliminates invading pathogens, restoring immune equilibrium without inflicting undue harm to the host. However, when a cascade of immunological reactions is triggered, the immune response can sometimes go into overdrive, potentially leading to harmful long-term effects or even death. The immune system is triggered mostly by infections, allergens, or medical interventions such as vaccination. This review examines how these immune triggers differ and why certain infections may dysregulate immune homeostasis, leading to inflammatory or allergic pathology and exacerbation of pre-existing conditions. However, many vaccines generate an optimal immune response and protect against the consequences of pathogen-induced immunological aggressiveness, and from a small number of unrelated pathogens and autoimmune diseases. Here, we propose an "immuno-wave" model describing a vaccine-induced "Goldilocks immunity", which leaves fine imprints of both pro-inflammatory and anti-inflammatory milieus, derived from both the innate and the adaptive arms of the immune system, in the body. The resulting balanced, 'quiet alert' state of the immune system may provide a jump-start in the defense against pathogens and any associated pathological inflammatory or allergic responses, allowing vaccines to go above and beyond their call of duty. In closing, we recommend formally investigating and reaping many of the secondary benefits of vaccines with appropriate clinical studies.
Collapse
Affiliation(s)
- Arun B Arunachalam
- Analytical Sciences, R&D Sanofi Vaccines, 1 Discovery Dr., Swiftwater, PA 18370, USA
| |
Collapse
|
3
|
Subburayalu J. Immune surveillance and humoral immune responses in kidney transplantation - A look back at T follicular helper cells. Front Immunol 2023; 14:1114842. [PMID: 37503334 PMCID: PMC10368994 DOI: 10.3389/fimmu.2023.1114842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/22/2023] [Indexed: 07/29/2023] Open
Abstract
T follicular helper cells comprise a specialized, heterogeneous subset of immune-competent T helper cells capable of influencing B cell responses in lymphoid tissues. In physiology, for example in response to microbial challenges or vaccination, this interaction chiefly results in the production of protecting antibodies and humoral memory. In the context of kidney transplantation, however, immune surveillance provided by T follicular helper cells can take a life of its own despite matching of human leukocyte antigens and employing the latest immunosuppressive regiments. This puts kidney transplant recipients at risk of subclinical and clinical rejection episodes with a potential risk for allograft loss. In this review, the current understanding of immune surveillance provided by T follicular helper cells is briefly described in physiological responses to contrast those pathological responses observed after kidney transplantation. Sensitization of T follicular helper cells with the subsequent emergence of detectable donor-specific human leukocyte antigen antibodies, non-human leukocyte antigen antibodies their implication for kidney transplantation and lessons learnt from other transplantation "settings" with special attention to antibody-mediated rejection will be addressed.
Collapse
Affiliation(s)
- Julien Subburayalu
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies (CRTD), Technische Universität Dresden, Dresden, Germany
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Cadar AN, Martin DE, Bartley JM. Targeting the hallmarks of aging to improve influenza vaccine responses in older adults. Immun Ageing 2023; 20:23. [PMID: 37198683 PMCID: PMC10189223 DOI: 10.1186/s12979-023-00348-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Abstract
Age-related declines in immune response pose a challenge in combating diseases later in life. Influenza (flu) infection remains a significant burden on older populations and often results in catastrophic disability in those who survive infection. Despite having vaccines designed specifically for older adults, the burden of flu remains high and overall flu vaccine efficacy remains inadequate in this population. Recent geroscience research has highlighted the utility in targeting biological aging to improve multiple age-related declines. Indeed, the response to vaccination is highly coordinated, and diminished responses in older adults are likely not due to a singular deficit, but rather a multitude of age-related declines. In this review we highlight deficits in the aged vaccine responses and potential geroscience guided approaches to overcome these deficits. More specifically, we propose that alternative vaccine platforms and interventions that target the hallmarks of aging, including inflammation, cellular senescence, microbiome disturbances, and mitochondrial dysfunction, may improve vaccine responses and overall immunological resilience in older adults. Elucidating novel interventions and approaches that enhance immunological protection from vaccination is crucial to minimize the disproportionate effect of flu and other infectious diseases on older adults.
Collapse
Affiliation(s)
- Andreia N Cadar
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Dominique E Martin
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Jenna M Bartley
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
| |
Collapse
|
5
|
Martin DE, Cadar AN, Panier H, Torrance BL, Kuchel GA, Bartley JM. The effect of metformin on influenza vaccine responses in nondiabetic older adults: a pilot trial. Immun Ageing 2023; 20:18. [PMID: 37131271 PMCID: PMC10152024 DOI: 10.1186/s12979-023-00343-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/24/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Aging is associated with progressive declines in immune responses leading to increased risk of severe infection and diminished vaccination responses. Influenza (flu) is a leading killer of older adults despite availability of seasonal vaccines. Geroscience-guided interventions targeting biological aging could offer transformational approaches to reverse broad declines in immune responses with aging. Here, we evaluated effects of metformin, an FDA approved diabetes drug and candidate anti-aging drug, on flu vaccination responses and markers of immunological resilience in a pilot and feasibility double-blinded placebo-controlled study. RESULTS Healthy older adults (non-diabetic/non-prediabetic, age: 74.4 ± 1.7 years) were randomized to metformin (n = 8, 1500 mg extended release/daily) or placebo (n = 7) treatment for 20 weeks and were vaccinated with high-dose flu vaccine after 10 weeks of treatment. Peripheral blood mononuclear cells (PBMCs), serum, and plasma were collected prior to treatment, immediately prior to vaccination, and 1, 5, and 10 weeks post vaccination. Increased serum antibody titers were observed post vaccination with no significant differences between groups. Metformin treatment led to trending increases in circulating T follicular helper cells post-vaccination. Furthermore, 20 weeks of metformin treatment reduced expression of exhaustion marker CD57 in circulating CD4 T cells. CONCLUSIONS Pre-vaccination metformin treatment improved some components of flu vaccine responses and reduced some markers of T cell exhaustion without serious adverse events in nondiabetic older adults. Thus, our findings highlight the potential utility of metformin to improve flu vaccine responses and reduce age-related immune exhaustion in older adults, providing improved immunological resilience in nondiabetic older adults.
Collapse
Affiliation(s)
- Dominique E Martin
- UConn Center On Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
- Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
| | - Andreia N Cadar
- UConn Center On Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
- Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
| | - Hunter Panier
- UConn Center On Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
- Department of Medicine, University of Connecticut School of Medicine, Farmington Avenue, Farmington, CT, 06030, USA
| | - Blake L Torrance
- UConn Center On Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
- Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
| | - George A Kuchel
- UConn Center On Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA
| | - Jenna M Bartley
- UConn Center On Aging, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA.
- Department of Immunology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, 860-679-8322, USA.
| |
Collapse
|
6
|
Ravichandran S, Erra-Diaz F, Karakaslar OE, Marches R, Kenyon-Pesce L, Rossi R, Chaussabel D, Pascual V, Palucka K, Paust S, Nahm MH, Kuchel GA, Banchereau J, Ucar D. Distinct baseline immune characteristics associated with responses to conjugated and unconjugated pneumococcal polysaccharide vaccines in older adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.16.23288531. [PMID: 37131707 PMCID: PMC10153339 DOI: 10.1101/2023.04.16.23288531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Pneumococcal infections cause serious illness and death among older adults. A capsular polysaccharide vaccine PPSV23 (Pneumovax®) and a conjugated polysaccharide vaccine PCV13 (Prevnar®) are used to prevent these infections, yet underlying responses, and baseline predictors remain unknown. We recruited and vaccinated 39 older adults (>60 years) with PPSV23 or PCV13. Both vaccines induced strong antibody responses at day 28 and similar plasmablast transcriptional signatures at day 10, however, their baseline predictors were distinct. Analyses of baseline flow cytometry and RNA-seq data (bulk and single cell) revealed a novel baseline phenotype that is specifically associated with weaker PCV13 responses, characterized by i) increased expression of cytotoxicity-associated genes and increased CD16+ NK frequency; ii) increased Th17 and decreased Th1 cell frequency. Men were more likely to display this cytotoxic phenotype and mounted weaker responses to PCV13 than women. Baseline expression levels of a distinct gene set was predictive of PPSV23 responses. This first precision vaccinology study for pneumococcal vaccine responses of older adults uncovered novel and distinct baseline predictors that might transform vaccination strategies and initiate novel interventions.
Collapse
Affiliation(s)
| | - Fernando Erra-Diaz
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- University of Buenos Aires, School of Medicine, Buenos Aires, Argentina #Current Address
| | - Onur E Karakaslar
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- Leiden University Medical Center (LUMC), Leiden, Netherlands #Current Address
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Lisa Kenyon-Pesce
- UConn Center on Aging, University of Connecticut, Farmington, Connecticut, USA
| | - Robert Rossi
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Damien Chaussabel
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Virginia Pascual
- Weill Cornell Medical College, Department of Pediatrics, NY, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Moon H Nahm
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut, Farmington, Connecticut, USA
| | - Jacques Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- Immunai, New York, NY, USA, #Current Address
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
- Institute for Systems Genomics, University of Connecticut Health Center, Farmington, Connecticut, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
7
|
Currenti J, Simmons J, Oakes J, Gaudieri S, Warren CM, Gangula R, Alves E, Ram R, Leary S, Armitage JD, Smith RM, Chopra A, Halasa NB, Pilkinton MA, Kalams SA. Tracking of activated cTfh cells following sequential influenza vaccinations reveals transcriptional profile of clonotypes driving a vaccine-induced immune response. Front Immunol 2023; 14:1133781. [PMID: 37063867 PMCID: PMC10095155 DOI: 10.3389/fimmu.2023.1133781] [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: 12/29/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction A vaccine against influenza is available seasonally but is not 100% effective. A predictor of successful seroconversion in adults is an increase in activated circulating T follicular helper (cTfh) cells after vaccination. However, the impact of repeated annual vaccinations on long-term protection and seasonal vaccine efficacy remains unclear. Methods In this study, we examined the T cell receptor (TCR) repertoire and transcriptional profile of vaccine-induced expanded cTfh cells in individuals who received sequential seasonal influenza vaccines. We measured the magnitude of cTfh and plasmablast cell activation from day 0 (d0) to d7 post-vaccination as an indicator of a vaccine response. To assess TCR diversity and T cell expansion we sorted activated and resting cTfh cells at d0 and d7 post-vaccination and performed TCR sequencing. We also single cell sorted activated and resting cTfh cells for TCR analysis and transcriptome sequencing. Results and discussion The percent of activated cTfh cells significantly increased from d0 to d7 in each of the 2016-17 (p < 0.0001) and 2017-18 (p = 0.015) vaccine seasons with the magnitude of cTfh activation increase positively correlated with the frequency of circulating plasmablast cells in the 2016-17 (p = 0.0001) and 2017-18 (p = 0.003) seasons. At d7 post-vaccination, higher magnitudes of cTfh activation were associated with increased clonality of cTfh TCR repertoire. The TCRs from vaccine-expanded clonotypes were identified and tracked longitudinally with several TCRs found to be present in both years. The transcriptomic profile of these expanded cTfh cells at the single cell level demonstrated overrepresentation of transcripts of genes involved in the type-I interferon pathway, pathways involved in gene expression, and antigen presentation and recognition. These results identify the expansion and transcriptomic profile of vaccine-induced cTfh cells important for B cell help.
Collapse
Affiliation(s)
- Jennifer Currenti
- School of Human Sciences, University of Western Australia, Crawley, WA, Australia
| | - Joshua Simmons
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jared Oakes
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Silvana Gaudieri
- School of Human Sciences, University of Western Australia, Crawley, WA, Australia
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Christian M. Warren
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rama Gangula
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eric Alves
- School of Human Sciences, University of Western Australia, Crawley, WA, Australia
| | - Ramesh Ram
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Shay Leary
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Jesse D. Armitage
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Rita M. Smith
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Natasha B. Halasa
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Mark A. Pilkinton
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Spyros A. Kalams
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
8
|
Picard E, Armstrong S, Andrew MK, Haynes L, Loeb M, Pawelec G, Kuchel GA, McElhaney JE, Verschoor CP. Markers of systemic inflammation are positively associated with influenza vaccine antibody responses with a possible role for ILT2(+)CD57(+) NK-cells. Immun Ageing 2022; 19:26. [PMID: 35619117 PMCID: PMC9134679 DOI: 10.1186/s12979-022-00284-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 05/15/2022] [Indexed: 02/06/2023]
Abstract
Background With increasing age, overall health declines while systemic levels of inflammatory mediators tend to increase. Although the underlying mechanisms are poorly understood, there is a wealth of data suggesting that this so-called “inflammaging” contributes to the risk of adverse outcomes in older adults. We sought to determine whether markers of systemic inflammation were associated with antibody responses to the seasonal influenza vaccine. Results Over four seasons, hemagglutination inhibition antibody titres and ex vivo bulk peripheral blood mononuclear cell (PBMC) responses to live influenza viruses assessed via interferon (IFN)-γ/interleukin (IL)-10 production, were measured pre- and 4-weeks post-vaccination in young adults (n = 79) and older adults randomized to standard- or high-dose inactivated vaccine (n = 612). Circulating tumour necrosis factor (TNF), interleukin (IL)-6 and C-reactive protein (CRP) were also measured pre-vaccination. Post-vaccination antibody titres were significantly associated with systemic inflammatory levels; specifically, IL-6 was positively associated with A/H3N2 titres in young adults (Cohen’s d = 0.36), and in older high-dose, but not standard-dose recipients, all systemic inflammatory mediators were positively associated with A/H1N1, A/H3N2 and B titres (d = 0.10–0.45). We further show that the frequency of ILT2(+)CD57(+) CD56-Dim natural killer (NK)-cells was positively associated with both plasma IL-6 and post-vaccination A/H3N2 titres in a follow-up cohort of older high-dose recipients (n = 63). Pathway analysis suggested that ILT2(+)CD57(+) Dim NK-cells mediated 40% of the association between IL-6 and A/H3N2 titres, which may be related to underlying participant frailty. Conclusions In summary, our data suggest a complex relationship amongst influenza vaccine responses, systemic inflammation and NK-cell phenotype in older adults, which depends heavily on age, vaccine dose and possibly overall health status. While our results suggest that “inflammaging” may increase vaccine immunogenicity in older adults, it is yet to be determined whether this enhancement contributes to improved protection against influenza disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12979-022-00284-x.
Collapse
|
9
|
Lai L, Rouphael N, Xu Y, Kabbani S, Beck A, Sherman A, Anderson EJ, Bellamy A, Weiss J, Cross K, Mulligan MJ. An Oil-in-Water adjuvant significantly increased influenza A/H7N9 split virus Vaccine-Induced circulating follicular helper T (cT FH) cells and antibody responses. Vaccine 2022; 40:7065-7072. [PMID: 36273986 DOI: 10.1016/j.vaccine.2022.09.041] [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: 07/28/2021] [Revised: 04/09/2022] [Accepted: 09/12/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Unadjuvanted A/H7N9 vaccines are poorly immunogenic. The immune response is improved with the addition of MF59, an oil-in-water adjuvant. However, the cellular immunologic responses of MF59-adjuvanted A/H7N9 vaccine are not fully understood. METHODS 37 participants were vaccinated with 2 doses of 2013 influenza A/H7N9 vaccine (at Days 1 and 21) with or without MF59 and enrolled in an immunology substudy. Responses were assessed at multiple timepoints (Days 0, 8, 21, 29, and 42) for hemagglutination inhibition (HAI) and neutralizing antibody (Neut) assays, memory B cell responses by enzyme-linked ImmunoSpot; circulating follicular helper T cells (cTFH) and CD4 + T cells by intracellular cytokine staining. RESULTS MF59-adjuvanted influenza A/H7N9 vaccine induced significantly higher hemagglutination inhibition (HAI) and neutralizing antibody (Neut) responses when compared to unadjuvanted vaccine. The adjuvanted vaccine elicited significantly higher levels of Inducible T-cell Co-Stimulator (ICOS) expression by CXCR3+CXCR5+CD4+ cTFH cells, compared to unadjuvanted vaccine. The magnitude of increase in cTFH cells (from baseline to Day 8) and in IL-21 expressing CD154+CD4+ T cells (from baseline to Days 8 and 21) correlated with HAI (at Day 29) and Neut antibody (at Days 8 and 29) titers. The increase in frequency of IL-21 expressing CD154+CD4+T cells (from baseline to Day 21) correlated with memory B cell frequency (at Day 42). CONCLUSION cTFH activation is associated with HAI and Neut responses in recipients of MF59-adjuvanted influenza A/H7N9 vaccine relative to unadjuvanted vaccine. Future studies should focus on optimizing the cTFH response and use cTFH as an early biomarker of serological response to vaccination. This trial was registered at clinicaltrials.gov, trial number NCT01938742.
Collapse
Affiliation(s)
- Lilin Lai
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030.
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Sarah Kabbani
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Allison Beck
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Amy Sherman
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| | - Evan J Anderson
- Departments of Pediatrics and Medicine, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate
| | - Abbie Bellamy
- EMMES Corporation, 401, North Washington Street, Suite 700, Rockville, MD 20850, USA
| | - Julia Weiss
- EMMES Corporation, 401, North Washington Street, Suite 700, Rockville, MD 20850, USA
| | - Kaitlyn Cross
- EMMES Corporation, 401, North Washington Street, Suite 700, Rockville, MD 20850, USA
| | - Mark J Mulligan
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University, 500, Irvin Court, Decatur GA 30030
| |
Collapse
|
10
|
Different antibody-associated autoimmune diseases have distinct patterns of T follicular cell dysregulation. Sci Rep 2022; 12:17638. [PMID: 36271118 PMCID: PMC9587230 DOI: 10.1038/s41598-022-21576-8] [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: 07/21/2022] [Accepted: 09/29/2022] [Indexed: 01/18/2023] Open
Abstract
Autoantibodies are produced within germinal centers (GC), in a process regulated by interactions between B, T follicular helper (Tfh), and T follicular regulatory (Tfr) cells. The GC dysregulation in human autoimmunity has been inferred from circulating cells, albeit with conflicting results due to diverse experimental approaches. We applied a consistent approach to compare circulating Tfr and Tfh subsets in patients with different autoimmune diseases. We recruited 97 participants, including 72 patients with Hashimoto's thyroiditis (HT, n = 18), rheumatoid arthritis (RA, n = 16), or systemic lupus erythematosus (SLE, n = 32), and 31 matched healthy donors (HD). We found that the frequency of circulating T follicular subsets differed across diseases. Patients with HT had an increased frequency of blood Tfh cells (p = 0.0215) and a reduced Tfr/Tfh ratio (p = 0.0338) when compared with HD. This was not observed in patients with systemic autoimmune rheumatic diseases (RA, SLE), who had a reduction in both Tfh (p = 0.0494 and p = 0.0392, respectively) and Tfr (p = 0.0003 and p = 0.0001, respectively) cells, resulting in an unchanged Tfr/Tfh ratio. Activated PD-1+ICOS+Tfh and CD4+PD-1+CXCR5-Tph cells were raised only in patients with SLE (p = 0.0022 and p = 0.0054), without association with disease activity. Our data suggest that GC dysregulation, assessed by T follicular subsets, is not uniform in human autoimmunity. Specific patterns of dysregulation may become potential biomarkers for disease and patient stratification.
Collapse
|
11
|
Cevirgel A, Shetty SA, Vos M, Nanlohy NM, Beckers L, Bijvank E, Rots N, van Beek J, Buisman A, van Baarle D. Identification of aging-associated immunotypes and immune stability as indicators of post-vaccination immune activation. Aging Cell 2022; 21:e13703. [PMID: 36081314 PMCID: PMC9577949 DOI: 10.1111/acel.13703] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/20/2022] [Accepted: 08/13/2022] [Indexed: 01/25/2023] Open
Abstract
Immunosenescence describes immune dysfunction observed in older individuals. To identify individuals at-risk for immune dysfunction, it is crucial to understand the diverse immune phenotypes and their intrinsic functional capabilities. We investigated immune cell subsets and variation in the aging population. We observed that inter-individual immune variation was associated with age and cytomegalovirus seropositivity. Based on the similarities of immune subset composition among individuals, we identified nine immunotypes that displayed different aging-associated immune signatures, which explained inter-individual variation better than age. Additionally, we correlated the immune subset composition of individuals over approximately a year as a measure of stability of immune parameters. Immune stability was significantly lower in immunotypes that contained aging-associated immune subsets and correlated with a circulating CD38 + CD4+ T follicular helper cell increase 7 days after influenza vaccination. In conclusion, immune stability is a feature of immunotypes and could be a potential indicator of post-vaccination cellular kinetics.
Collapse
Affiliation(s)
- Alper Cevirgel
- Center for Infectious Disease ControlNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands,Department of Medical Microbiology and Infection preventionVirology 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 preventionVirology 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 preventionVirology and Immunology Research GroupUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
12
|
Yu D, Walker LSK, Liu Z, Linterman MA, Li Z. Targeting T FH cells in human diseases and vaccination: rationale and practice. Nat Immunol 2022; 23:1157-1168. [PMID: 35817844 DOI: 10.1038/s41590-022-01253-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022]
Abstract
The identification of CD4+ T cells localizing to B cell follicles has revolutionized the knowledge of how humoral immunity is generated. Follicular helper T (TFH) cells support germinal center (GC) formation and regulate clonal selection and differentiation of memory and antibody-secreting B cells, thus controlling antibody affinity maturation and memory. TFH cells are essential in sustaining protective antibody responses necessary for pathogen clearance in infection and vaccine-mediated protection. Conversely, aberrant and excessive TFH cell responses mediate and sustain pathogenic antibodies to autoantigens, alloantigens, and allergens, facilitate lymphomagenesis, and even harbor viral reservoirs. TFH cell generation and function are determined by T cell antigen receptor (TCR), costimulation, and cytokine signals, together with specific metabolic and survival mechanisms. Such regulation is crucial to understanding disease pathogenesis and informing the development of emerging therapies for disease or novel approaches to boost vaccine efficacy.
Collapse
Affiliation(s)
- Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia. .,Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Division of Infection & Immunity, University College London, Royal Free Campus, London, UK
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Zhanguo Li
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
13
|
Nicoli F, Mantelli B, Gallerani E, Telatin V, Squarzon L, Masiero S, Gavioli R, Palù G, Barzon L, Caputo A. Effects of the age of vaccination on the humoral responses to a human papillomavirus vaccine. NPJ Vaccines 2022; 7:37. [PMID: 35292655 PMCID: PMC8924199 DOI: 10.1038/s41541-022-00458-0] [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: 05/24/2021] [Accepted: 02/11/2022] [Indexed: 11/11/2022] Open
Abstract
Adult vaccination programs are receiving increasing attention however, little is known regarding the impact of age on the maintenance of the immune response. We investigated this issue in the context of a human papillomavirus (HPV) vaccination program collecting real-world data on the durability of humoral immunity in 315 female subjects stratified according to vaccination age (adolescents and adults) and sampled at early or late time points after the last vaccine dose. HPV-specific IgGs, but not memory B cells, were induced and maintained at higher levels in subjects vaccinated during adolescence. Nonetheless, antibody functions waned over time to a similar degree in adolescents and adults. To shed light on this phenomena, we analyzed quantitative and qualitative properties of lymphocytes. Similar biochemical features were observed between B-cell subsets from individuals belonging to the two age groups. Long term humoral responses toward vaccines administered at an earlier age were comparably maintained between adolescents and adults. The percentages of naïve B and CD4+ T cells were significantly higher in adolescents, and the latter directly correlated with IgG titers against 3 out of 4 HPV types. Our results indicate that age-specific HPV vaccine responsiveness is mostly due to quantitative differences of immune cell precursors rather than qualitative defects in B cells. In addition, our results indicate that adults also have a good humoral immunogenic profile, suggesting that their inclusion in catch-up programmes is desirable.
Collapse
Affiliation(s)
- Francesco Nicoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Barbara Mantelli
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Eleonora Gallerani
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Valentina Telatin
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Laura Squarzon
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Serena Masiero
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Riccardo Gavioli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Antonella Caputo
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy.
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy.
| |
Collapse
|
14
|
Law H, Mach M, Howe A, Obeid S, Milner B, Carey C, Elfis M, Fsadni B, Ognenovska K, Phan TG, Carey D, Xu Y, Venturi V, Zaunders J, Kelleher AD, Munier CML. Early expansion of CD38+ICOS+ GC Tfh in draining lymph nodes during influenza vaccination immune response. iScience 2022; 25:103656. [PMID: 35028536 PMCID: PMC8741621 DOI: 10.1016/j.isci.2021.103656] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/22/2021] [Accepted: 12/14/2021] [Indexed: 01/23/2023] Open
Abstract
T follicular helper (Tfh) cells provide critical help to B cells during the germinal center (GC) reaction to facilitate generation of protective humoral immunity. Accessing the human lymph node (LN) to study the commitment of CD4 T cells to GC Tfh cell differentiation during in vivo vaccine responses is difficult. We used ultrasound guided fine needle biopsy to monitor recall responses in axillary LNs to seasonal influenza vaccination in healthy volunteers. Specific expansion of GC cell subsets occurred exclusively within draining LNs five days postvaccination. Draining LN GC Tfh and precursor-Tfh cells express higher levels of CD38, ICOS, and Ki67, indicating they were significantly more activated, motile, and proliferating, compared to contralateral LN cells. These observations provide insight into the early expansion phase of the human Tfh lineage within LNs during a vaccine induced memory response and highlights early LN immune responses may not be reflected in the periphery. Early response to influenza vaccine is characterized by expansion of GC cell subsets Specific expansion of CD38+ ICOS+ GC Tfh and Pre-Tfh occurs in draining LNs only Activated GC Tfh and Pre-Tfh are also proliferating, expressing high levels of Ki67 Correlation between activated Pre-Tfh and activated c-Tfh suggests a potential origin
Collapse
Affiliation(s)
- Hannah Law
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia
| | - Melanie Mach
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia.,The University of Sydney, Sydney 2006, NSW, Australia
| | - Annett Howe
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia
| | - Solange Obeid
- St Vincent's Hospital Sydney, Sydney 2010, NSW, Australia
| | - Brad Milner
- St Vincent's Hospital Sydney, Sydney 2010, NSW, Australia
| | - Cate Carey
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia
| | - Maxine Elfis
- St Vincent's Hospital Sydney, Sydney 2010, NSW, Australia
| | - Bertha Fsadni
- St Vincent's Centre for Applied Medical Research (AMR), Sydney 2010, NSW, Australia
| | | | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney 2010, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney 2010, NSW, Australia
| | - Diane Carey
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia
| | - Yin Xu
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia
| | - Vanessa Venturi
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia
| | - John Zaunders
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia.,St Vincent's Centre for Applied Medical Research (AMR), Sydney 2010, NSW, Australia
| | - Anthony D Kelleher
- The Kirby Institute, UNSW Sydney, Sydney 2052, NSW, Australia.,St Vincent's Hospital Sydney, Sydney 2010, NSW, Australia.,St Vincent's Centre for Applied Medical Research (AMR), Sydney 2010, NSW, Australia
| | | |
Collapse
|
15
|
Juno JA, Hill DL. T follicular helper cells and their impact on humoral responses during pathogen and vaccine challenge. Curr Opin Immunol 2021; 74:112-117. [PMID: 34861545 DOI: 10.1016/j.coi.2021.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
Abstract
T follicular helper (Tfh) cells are essential for the establishment, maintenance and output of the germinal centre (GC) response. The transient nature of this response, and its location within secondary lymphoid tissues have hampered our understanding of this critical cell type, particularly in humans. A counterpart of GC Tfh cells in peripheral blood has enabled recent discoveries in disease and vaccination settings, while direct sampling of lymph nodes provides exciting new avenues to study GC responses directly in vivo. Tfh differentiation is shaped by the cytokine milieu during inflammation, vaccination and with age, and disease-specific patterns are emerging. An improved understanding of how to support a Tfh response remains key to enhancing vaccine immunity across the lifespan.
Collapse
Affiliation(s)
- Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne 3000, Victoria, Australia.
| | - Danika L Hill
- Department of Immunology and Pathology, Monash University, 89 Commercial Rd., Melbourne 3004, Victoria, Australia.
| |
Collapse
|
16
|
Wild K, Smits M, Killmer S, Strohmeier S, Neumann-Haefelin C, Bengsch B, Krammer F, Schwemmle M, Hofmann M, Thimme R, Zoldan K, Boettler T. Pre-existing immunity and vaccine history determine hemagglutinin-specific CD4 T cell and IgG response following seasonal influenza vaccination. Nat Commun 2021; 12:6720. [PMID: 34795301 PMCID: PMC8602312 DOI: 10.1038/s41467-021-27064-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Effectiveness of seasonal influenza vaccination varies between individuals and might be affected by vaccination history among other factors. Here we show, by monitoring frequencies of CD4 T cells specific to the conserved hemagglutinin epitope HA118-132 and titres of IgG against the corresponding recombinant hemagglutinin protein, that antigen-specific CD4 T cell and antibody responses are closely linked to pre-existing immunity and vaccine history. Upon immunization, a strong early reaction is observed in all vaccine naïve participants and also in vaccine experienced individuals who have not received the respective seasonal vaccine in the previous year. This response is characterized by HA118-132 specific CD4 T cells with a follicular helper T cell phenotype and by ascending titers of hemagglutinin-specific antibodies from baseline to day 28 following vaccination. This trend was observed in only a proportion of those participants who received the seasonal vaccine the year preceding the study. Regardless of history, levels of pre-existing antibodies and CD127 expression on CD4 T cells at baseline were the strongest predictors of robust early response. Thus, both pre-existing immunity and vaccine history contribute to the response to seasonal influenza vaccines.
Collapse
Affiliation(s)
- Katharina Wild
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Pharmacy, University of Freiburg, Freiburg, Germany
| | - Maike Smits
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Saskia Killmer
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Shirin Strohmeier
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christoph Neumann-Haefelin
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bertram Bengsch
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Schwemmle
- Institute of Virology, Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katharina Zoldan
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Boettler
- Department of Medicine II, Medical Center - University of Freiburg, Freiburg, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
17
|
Cohen CA, Li APY, Hachim A, Hui DSC, Kwan MYW, Tsang OTY, Chiu SS, Chan WH, Yau YS, Kavian N, Ma FNL, Lau EHY, Cheng SMS, Poon LLM, Peiris M, Valkenburg SA. SARS-CoV-2 specific T cell responses are lower in children and increase with age and time after infection. Nat Commun 2021; 12:4678. [PMID: 34326343 PMCID: PMC8322064 DOI: 10.1038/s41467-021-24938-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 infection of children leads to a mild illness and the immunological differences with adults are unclear. Here, we report SARS-CoV-2 specific T cell responses in infected adults and children and find that the acute and memory CD4+ T cell responses to structural SARS-CoV-2 proteins increase with age, whereas CD8+ T cell responses increase with time post-infection. Infected children have lower CD4+ and CD8+ T cell responses to SARS-CoV-2 structural and ORF1ab proteins when compared with infected adults, comparable T cell polyfunctionality and reduced CD4+ T cell effector memory. Compared with adults, children have lower levels of antibodies to β-coronaviruses, indicating differing baseline immunity. Total T follicular helper responses are increased, whilst monocyte numbers are reduced, indicating rapid adaptive co-ordination of the T and B cell responses and differing levels of inflammation. Therefore, reduced prior β-coronavirus immunity and reduced T cell activation in children might drive milder COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Carolyn A Cohen
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Athena P Y Li
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - David S C Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Mike Y W Kwan
- Department of Paediatric and Adolescent Medicine, Hong Kong Hospital Authority Infectious Disease Center, Princess Margaret Hospital, Hong Kong, SAR, China
| | - Owen T Y Tsang
- Infectious Diseases Centre, Princess Margaret Hospital, Hospital Authority of Hong Kong, Hong Kong SAR, China
| | - Susan S Chiu
- Department of Paediatric and Adolescent Medicine, The University of Hong Kong and Queen Mary Hospital, Hospital Authority of Hong Kong, Hong Kong SAR, China
| | - Wai Hung Chan
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Hong Kong SAR, China
| | - Yat Sun Yau
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Hong Kong SAR, China
| | - Niloufar Kavian
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Fionn N L Ma
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Eric H Y Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Samuel M S Cheng
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Leo L M Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
18
|
The metabolic hormone leptin promotes the function of T FH cells and supports vaccine responses. Nat Commun 2021; 12:3073. [PMID: 34031386 PMCID: PMC8144586 DOI: 10.1038/s41467-021-23220-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
Follicular helper T (TFH) cells control antibody responses by supporting antibody affinity maturation and memory formation. Inadequate TFH function has been found in individuals with ineffective responses to vaccines, but the mechanism underlying TFH regulation in vaccination is not understood. Here, we report that lower serum levels of the metabolic hormone leptin associate with reduced vaccine responses to influenza or hepatitis B virus vaccines in healthy populations. Leptin promotes mouse and human TFH differentiation and IL-21 production via STAT3 and mTOR pathways. Leptin receptor deficiency impairs TFH generation and antibody responses in immunisation and infection. Similarly, leptin deficiency induced by fasting reduces influenza vaccination-mediated protection for the subsequent infection challenge, which is mostly rescued by leptin replacement. Our results identify leptin as a regulator of TFH cell differentiation and function and indicate low levels of leptin as a risk factor for vaccine failure. T follicular helper (TFH) cell numbers are increased after vaccination and fewer of these cells might result in reduced vaccine responses. Here the authors show in mice and humans that leptin promotes TFH differentiation and that low leptin levels can impair TFH response to vaccines and virus protection in mice.
Collapse
|
19
|
Topham DJ, DeDiego ML, Nogales A, Sangster MY, Sant A. Immunity to Influenza Infection in Humans. Cold Spring Harb Perspect Med 2021; 11:a038729. [PMID: 31871226 PMCID: PMC7919402 DOI: 10.1101/cshperspect.a038729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review discusses the human immune responses to influenza infection with some insights from studies using animal models, such as experimental infection of mice. Recent technological advances in the study of human immune responses have greatly added to our knowledge of the infection and immune responses, and therefore much of the focus is on recent studies that have moved the field forward. We consider the complexity of the adaptive response generated by many sequential encounters through infection and vaccination.
Collapse
Affiliation(s)
- David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Marta L DeDiego
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Cientificas, 28049 Madrid, Spain
| | - Aitor Nogales
- Instituto Nacional de Investigación y Tecnologia Agraria y Ailmentaria, 28040 Madrid, Spain
| | - Mark Y Sangster
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Andrea Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
20
|
Protein/AS01 B vaccination elicits stronger, more Th2-skewed antigen-specific human T follicular helper cell responses than heterologous viral vectors. CELL REPORTS MEDICINE 2021; 2:100207. [PMID: 33763653 PMCID: PMC7974546 DOI: 10.1016/j.xcrm.2021.100207] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/12/2020] [Accepted: 01/29/2021] [Indexed: 12/20/2022]
Abstract
Interactions between B cells and CD4+ T follicular helper (Tfh) cells are key determinants of humoral responses. Using samples from clinical trials performed with the malaria vaccine candidate antigen Plasmodium falciparum merozoite protein (PfRH5), we compare the frequency, phenotype, and gene expression profiles of PfRH5-specific circulating Tfh (cTfh) cells elicited by two leading human vaccine delivery platforms: heterologous viral vector prime boost and protein with AS01B adjuvant. We demonstrate that the protein/AS01B platform induces a higher-magnitude antigen-specific cTfh cell response and that this correlates with peak anti-PfRH5 IgG concentrations, frequency of PfRH5-specific memory B cells, and antibody functionality. Furthermore, our data indicate a greater Th2/Tfh2 skew within the polyfunctional response elicited following vaccination with protein/AS01B as compared to a Th1/Tfh1 skew with viral vectors. These data highlight the impact of vaccine platform on the cTfh cell response driving humoral immunity, associating a high-magnitude, Th2-biased cTfh response with potent antibody production. CD4 Tfh comparison in malaria vaccine trials using leading human vaccine platforms Protein/AS01B drives stronger antigen-specific Tfh responses than viral vectors Greater T(f)h2 skewing of antigen-specific CD4 T cells in protein/AS01B vaccinees Antigen-specific CD4 T(fh) cell parameters correlate with functional antibody
Collapse
|
21
|
Cohen CA, Li APY, Hachim A, Hui DSC, Kwan MYW, Tsang OTY, Chiu SS, Chan WH, Yau YS, Kavian N, Ma FNL, Lau EHY, Cheng SMS, Poon LLM, Peiris JSM, Valkenburg SA. SARS-CoV-2 specific T cell responses are lower in children and increase with age and time after infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.02.21250988. [PMID: 33564773 PMCID: PMC7872365 DOI: 10.1101/2021.02.02.21250988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
SARS-CoV-2 infection of children leads to a mild illness and the immunological differences with adults remains unclear. We quantified the SARS-CoV-2 specific T cell responses in adults and children (<13 years of age) with RT-PCR confirmed asymptomatic and symptomatic infection for long-term memory, phenotype and polyfunctional cytokines. Acute and memory CD4+ T cell responses to structural SARS-CoV-2 proteins significantly increased with age, whilst CD8+ T cell responses increased with time post infection. Infected children had significantly lower CD4+ and CD8+ T cell responses to SARS-CoV-2 structural and ORF1ab proteins compared to infected adults. SARS-CoV-2-specific CD8+ T cell responses were comparable in magnitude to uninfected negative adult controls. In infected adults CD4+ T cell specificity was skewed towards structural peptides, whilst children had increased contribution of ORF1ab responses. This may reflect differing T cell compartmentalisation for antigen processing during antigen exposure or lower recruitment of memory populations. T cell polyfunctional cytokine production was comparable between children and adults, but children had a lower proportion of SARS-CoV-2 CD4+ T cell effector memory. Compared to adults, children had significantly lower levels of antibodies to β-coronaviruses, indicating differing baseline immunity. Total T follicular helper responses was increased in children during acute infection indicating rapid co-ordination of the T and B cell responses. However total monocyte responses were reduced in children which may be reflective of differing levels of inflammation between children and adults. Therefore, reduced prior β-coronavirus immunity and reduced activation and recruitment of de novo responses in children may drive milder COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Carolyn A Cohen
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Athena PY Li
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David SC Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mike YW Kwan
- Department of Paediatric and Adolescent Medicine, Hong Kong Hospital Authority Infectious Disease Center, Princess Margaret Hospital, Special Administrative Region of Hong Kong, China
| | - Owen TY Tsang
- Infectious Diseases Centre, Princess Margaret Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Susan S Chiu
- Department of Paediatric and Adolescent Medicine, The University of Hong Kong and Queen Mary Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Wai Hung Chan
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Yat Sun Yau
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Niloufar Kavian
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Fionn NL Ma
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Eric HY Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Samuel MS Cheng
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Leo LM Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - JS Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
22
|
Caldera F, Mercer M, Samson SI, Pitt JM, Hayney MS. Influenza vaccination in immunocompromised populations: Strategies to improve immunogenicity. Vaccine 2021; 39 Suppl 1:A15-A23. [PMID: 33422377 DOI: 10.1016/j.vaccine.2020.11.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/22/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
Immunocompromised individuals are at high risk of severe illness and complications from influenza infection. For this reason, immunization using inactivated influenza vaccines is recommended for transplant patients, individuals receiving immunosuppressant treatments, and other persons with immunodeficiency. However, these immunocompromised populations are more likely to have lower and non-protective responses to annual vaccination with a standard influenza vaccine. Here, we review strategies aimed to improve the immunogenicity of influenza vaccines in immunocompromised populations. The different strategies employed have included adjuvanted vaccines, high-dose vaccines, booster doses, intradermal vaccination, and temporary discontinuation of immunosuppressant treatment regimens. High-dose trivalent, inactivated, split-virus influenza vaccine (IIV3-HD) is so far one of the leading strategies for improving vaccine responses in HIV patients, transplant patients, and persons receiving immunosuppressant therapies for inflammatory diseases. Several studies in these populations have shown stronger humoral responses with IIV3-HD than existing standard-dose trivalent vaccine, and comparable safety. Accordingly, some scientific societies have stated that high-dose influenza vaccine could be a preferred option for immunocompromised patients. However, larger randomized controlled studies are needed to validate relative immunogenicity and safety of IIV3-HD and other enhanced vaccines and vaccination strategies in immunocompromised individuals.
Collapse
Affiliation(s)
- Freddy Caldera
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | | | | | | | - Mary S Hayney
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
23
|
Tfh Cells in Health and Immunity: Potential Targets for Systems Biology Approaches to Vaccination. Int J Mol Sci 2020; 21:ijms21228524. [PMID: 33198297 PMCID: PMC7696930 DOI: 10.3390/ijms21228524] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
T follicular helper (Tfh) cells are a specialised subset of CD4+ T cells that play a significant role in the adaptive immune response, providing critical help to B cells within the germinal centres (GC) of secondary lymphoid organs. The B cell receptors of GC B cells undergo multiple rounds of somatic hypermutation and affinity maturation within the GC response, a process dependent on cognate interactions with Tfh cells. B cells that receive sufficient help from Tfh cells form antibody-producing long-lived plasma and memory B cells that provide the basis of decades of effective and efficient protection and are considered the gold standard in correlates of protection post-vaccination. However, the T cell response to vaccination has been understudied, and over the last 10 years, exponential improvements in the technological underpinnings of sampling techniques, experimental and analytical tools have allowed multidisciplinary characterisation of the role of T cells and the immune system as a whole. Of particular interest to the field of vaccinology are GCs and Tfh cells, representing a unique target for improving immunisation strategies. Here, we discuss recent insights into the unique journey of Tfh cells from thymus to lymph node during differentiation and their role in the production of high-quality antibody responses as well as their journey back to the periphery as a population of memory cells. Further, we explore their function in health and disease and the power of next-generation sequencing techniques to uncover their potential as modulators of vaccine-induced immunity.
Collapse
|
24
|
Yin M, Xiong Y, Huang L, Liu G, Yu Z, Zhao Y, Zhao J, Zhang Y, Lian T, Huang J, Liang D, Zeng J, Ni J. Circulating follicular helper T cells and subsets are associated with immune response to hepatitis B vaccination. Hum Vaccin Immunother 2020; 17:566-574. [PMID: 32614645 DOI: 10.1080/21645515.2020.1775457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Around 5-10% of healthy vaccinees lack or produce an inadequate antibody response following receipt of a standard hepatitis B vaccination regimen. Studying immune response to hepatitis B vaccination could promote researches of immunological events contributing to this poor response. To address this, we investigated follicular helper T (Tfh) cells and firstly demonstrated similar kinetics between circulating Tfh (cTfh) cells and Tfh cells derived from mice spleen after hepatitis B vaccination. And cTfh cells were positively associated with anti-HBs at one week after vaccination (D7). Furthermore, we found PBMCs stimulated by HBsAg showed preferential activation of CXCR3- Tfh cells subsets in vitro. The expression of transcription factor BCL6 in CD4+ T cell significantly differed between D7 and four weeks after vaccination (D28). However, dynamic curve of CD19+ B cells tended to rise then fall but no significant trends were observed. Our findings revealed a decrease in cTfh cells and subset skewing contribute to reduced antibody responses in immune response to hepatitis B vaccination, which indicated the importance of Tfh cell in facilitating the optimization of vaccine efficacy.
Collapse
Affiliation(s)
- Mingjuan Yin
- Department of Preventive Medicine, Guangdong Medical University , Dongguan, China.,Maternal and Child Research Institute, Women and Children's Hospital Affiliated to Guangdong Medical University (Shunde District Maternal and Child Health Hospital) , Foshan, China
| | - Yongzhen Xiong
- School Clinic, Guangdong Medical University , Dongguan, China
| | - Lingfeng Huang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Gang Liu
- Department of Immunization Program, Shenzhen Center for Disease Control and Prevention , Shenzhen, China
| | - Zuwei Yu
- Public Health Office, Dalang Town Community Health Service Center , Dongguan, China
| | - Yi Zhao
- Microbiology and Immunology, Guangdong Medical University , Dongguan, China
| | - Jie Zhao
- Neonatal Department, Shenzhen Maternal and Child Health Hospital Affiliated to Southern Medical University , Shenzhen, China
| | - Yan Zhang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Tingyu Lian
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Jingxiao Huang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - DongMei Liang
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - JinMei Zeng
- Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| | - Jindong Ni
- Maternal and Child Research Institute, Women and Children's Hospital Affiliated to Guangdong Medical University (Shunde District Maternal and Child Health Hospital) , Foshan, China.,Department of Epidemiology and Biostatistics, Guangdong Medical University , Dongguan, China
| |
Collapse
|
25
|
Li Y, Guptill JT, Russo MA, Howard JF, Massey JM, Juel VC, Hobson-Webb LD, Emmett D, Chopra M, Raja S, Liu W, Yi JS. Imbalance in T follicular helper cells producing IL-17 promotes pro-inflammatory responses in MuSK antibody positive myasthenia gravis. J Neuroimmunol 2020; 345:577279. [PMID: 32497931 DOI: 10.1016/j.jneuroim.2020.577279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/08/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022]
Abstract
A detailed understanding of the role of Tfh cells in MuSK-antibody positive myasthenia gravis (MuSK-MG) is lacking. We characterized phenotype and function of Tfh cells in MuSK-MG patients and controls. We found similar overall Tfh and follicular regulatory (Tfr) T cell frequencies in MuSK-MG and healthy controls, but MuSK-MG patients exhibited higher frequencies of Tfh17 cells and a higher ratio of Tfh:Tfr cells. These results suggest imbalanced Tfh cell regulation, further supported by increased frequencies of CD4 T cells co-producing IL-21/IL-17 and IL-17/IFN-γ, and increased Tfh-supported IgG production. These results support a role for Tfh cell dysregulation in MuSK-MG immunopathology.
Collapse
Affiliation(s)
- Yingkai Li
- Department of Neurology, Duke University Medical Center, Durham, NC, USA; Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jeffrey T Guptill
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Melissa A Russo
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - James F Howard
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janice M Massey
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Vern C Juel
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Lisa D Hobson-Webb
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Doug Emmett
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Manisha Chopra
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shruti Raja
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Weibin Liu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - John S Yi
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
26
|
Isakova-Sivak I, Grigorieva E, Rudenko L. Insights into current clinical research on the immunogenicity of live attenuated influenza vaccines. Expert Rev Vaccines 2020; 19:43-55. [PMID: 31903816 DOI: 10.1080/14760584.2020.1711056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: Live attenuated influenza vaccines (LAIVs) have been in use for more than three decades and are now licensed in many countries. There is evidence that LAIVs can have greater efficacy than inactivated influenza vaccines, especially against mismatched influenza, however, in recent years, a number of trials have found a lack of LAIV efficacy, mainly in relation to the H1N1 virus.Areas covered: In this review, we summarize the results of clinical research published in the past 5 years on the immunogenicity of LAIVs, with special attention to the mechanisms of establishing protective immunity and some factors that may influence immunogenicity and efficacy.Expert opinion: A number of recent clinical studies confirmed that the immune responses to LAIVs are multifaceted, involving different immune mechanisms. These trials suggest that the intrinsic replicative properties of each LAIV component should be taken into account, and the precise effects of adding a fourth vaccine strain to trivalent LAIV formulations are still to be identified. In addition, new data are emerging regarding the impact of pre-vaccination conditions, such as preexisting immunity or concurrent asymptomatic viral and bacterial respiratory infections, on LAIV immunogenicity, suggesting the importance of monitoring them during clinical trials or vaccination campaigns.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Elena Grigorieva
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
27
|
A Multiple Antigen Peptide Vaccine Containing CD4 + T Cell Epitopes Enhances Humoral Immunity against Trichinella spiralis Infection in Mice. J Immunol Res 2020; 2020:2074803. [PMID: 32377530 PMCID: PMC7199560 DOI: 10.1155/2020/2074803] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/06/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022] Open
Abstract
Multiepitope peptide vaccine has some advantages over traditional recombinant protein vaccine due to its easy and fast production and possible inclusion of multiple protective epitopes of pathogens. However, it is usually poorly immunogenic and needs to conjugate to a large carrier protein. Peptides conjugated to a central lysine core to form multiple antigen peptides (MAPs) will increase the immunogenicity of peptide vaccine. In this study, we constructed a MAP consisting of CD4+ T cell and B cell epitopes of paramyosin (Pmy) of Trichinella spiralis (Ts-Pmy), which has been proved to be a good vaccine candidate in our previous work. The immunogenicity and induced protective immunity of MAP against Trichinella spiralis (T. spiralis) infection were evaluated in mice. We demonstrated that mice immunized with MAP containing CD4+ T cell and B cell epitopes (MAP-TB) induced significantly higher protection against the challenge of T. spiralis larvae (35.5% muscle larva reduction) compared to the MAP containing B cell epitope alone (MAP-B) with a 12.4% muscle larva reduction. The better protection induced by immunization of MAP-TB was correlated with boosted antibody titers (both IgG1 and IgG2a) and mixed Th1/Th2 cytokine production secreted by the splenocytes of immunized mice. Further flow cytometry analysis of lymphocytes in spleens and draining lymph nodes demonstrated that mice immunized with MAP-TB specifically enhanced the generation of T follicular helper (Tfh) cells and germinal center (GC) B cells, while inhibiting follicular regulatory CD4+ T (Tfr) cells and regulatory T (Treg) cells. Immunofluorescence staining of spleen sections also confirmed that MAP-TB vaccination enhanced the formation of GCs. Our results suggest that CD4+ T cell epitope of Ts-Pmy is crucial in vaccine component for inducing better protection against T. spiralis infection.
Collapse
|
28
|
Biswas A, Chakrabarti AK, Dutta S. Current challenges: from the path of “original antigenic sin” towards the development of universal flu vaccines. Int Rev Immunol 2019; 39:21-36. [DOI: 10.1080/08830185.2019.1685990] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Asim Biswas
- Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Alok K. Chakrabarti
- Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Shanta Dutta
- Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| |
Collapse
|
29
|
Koutsakos M, Nguyen THO, Kedzierska K. With a Little Help from T Follicular Helper Friends: Humoral Immunity to Influenza Vaccination. THE JOURNAL OF IMMUNOLOGY 2019; 202:360-367. [PMID: 30617117 DOI: 10.4049/jimmunol.1800986] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/12/2018] [Indexed: 12/24/2022]
Abstract
Influenza remains a global and unpredictable threat. Annual vaccination against influenza A and B viruses promotes the induction of Abs and memory B cells, which can provide strain-specific protection against subsequent infections. The formation of effective memory B cell and Ab responses is highly dependent on the germinal center reaction, a well-orchestrated process involving B cells and a specialized CD4+ T cell subset called T follicular helper (Tfh) cells. As Tfh cells predominantly reside within B cell follicles in secondary lymphoid organs, they are challenging to study in humans. Recent identification of a circulating counterpart of Tfh cells has allowed us to better understand the contribution of these circulating Tfh cells during human immune responses. In this article, we summarize the role of human Tfh cells during humoral immune responses and discuss the contribution of Tfh cells in promoting immunity to influenza viruses in healthy cohorts and high-risk groups.
Collapse
Affiliation(s)
- Marios Koutsakos
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville 3010, Victoria, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville 3010, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Parkville 3010, Victoria, Australia
| |
Collapse
|
30
|
Kohut M. Exercise and psychoneuroimmunology. Curr Opin Behav Sci 2019. [DOI: 10.1016/j.cobeha.2019.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
31
|
Noh JY, Jang YS, Lee SN, Choi MJ, Yoon JG, Yu DH, Song JY, Cheong HJ, Kim WJ. Randomized, single-blind, active-controlled phase I clinical trial to evaluate the immunogenicity and safety of GC3114 (high-dose, quadrivalent influenza vaccine) in healthy adults. Vaccine 2019; 37:5171-5176. [PMID: 31377075 DOI: 10.1016/j.vaccine.2019.07.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 11/27/2022]
Abstract
Influenza is a major medically attended respiratory illness. The impact of influenza on morbidity and mortality is particularly high in the elderly. Immunosenescence attenuates the immune response of influenza vaccine in the elderly. High-dose influenza vaccine contains 60 μg of hemagglutinin per strain, four times more compared with standard-dose (SD) influenza vaccine. This study is a phase I clinical trial investigating the immunogenicity and safety of the GC3114, high-dose, quadrivalent inactivated influenza vaccine (HD-QIV) in healthy adults aged 19-64 years during the 2017-2018 season. Seroprotection rates of HD-QIV were 100.0% for A/H1N1, 96.67% for A/H3N2, 83.33% for B/Yamagata, and 96.67% for B/Victoria. Seroconversion rate for A/H1N1, A/H3N2, B/Yamagata, and B/Victoria strains were 86.67%, 90.0%, 53.33%, and 53.33%, respectively, in the HD-QIV group. The post-/pre-vaccination geometric mean titer ratio (GMTR) was 15.28 for A/H1N1, 8.19 for A/H3N2, 3.56 for B/Yamagata, and 3.03 for B/Victoria in the HD-QIV group. Seroconversion rate and post-/pre-vaccination GMTR for A/H3N2 were significantly higher in the HD-QIV group than in the SD-QIV group (control). No serious adverse events were reported. In conclusion, GC3114 was safe, well-tolerated, and immunogenic in healthy adults. Clinical Trials Identifier: NCT03357263.
Collapse
Affiliation(s)
- Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea; Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ye Seul Jang
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Saem Na Lee
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Min Joo Choi
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea; Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jin Gu Yoon
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Du Hyeon Yu
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea; Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea; Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo Joo Kim
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea; Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Fox A, Quinn KM, Subbarao K. Extending the Breadth of Influenza Vaccines: Status and Prospects for a Universal Vaccine. Drugs 2019; 78:1297-1308. [PMID: 30088204 DOI: 10.1007/s40265-018-0958-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite the widespread use of seasonal influenza vaccines, there is urgent need for a universal influenza vaccine to provide broad, long-term protection. A number of factors underpin this urgency, including threats posed by zoonotic and pandemic influenza A viruses, suboptimal effectiveness of seasonal influenza vaccines, and concerns surrounding the effects of annual vaccination. In this article, we discuss approaches that are being investigated to increase influenza vaccine breadth, which are near-term, readily achievable approaches to increase the range of strains recognized within a subtype, or longer-term more challenging approaches to produce a truly universal influenza vaccine. Adjuvanted and neuraminidase-optimized vaccines are emerging as the most feasible and promising approaches to extend protection to cover a broader range of strains within a subtype. The goal of developing a universal vaccine has also been advanced with the design of immunogenic influenza HA-stem constructs that induce broadly neutralizing antibodies. However, these constructs are not yet sufficiently immunogenic to induce lasting universal immunity in humans. Advances in understanding how T cells mediate protection, and how viruses are packaged, have facilitated the rationale design and delivery of replication-incompetent virus vaccines that induce broad protection mediated by lung-resident memory T cells. While the lack of clear mechanistic correlates of protection, other than haemagglutination-inhibiting antibodies, remains an impediment to further advancing novel influenza vaccines, the pressing need for such a vaccine is supporting development of highly innovative and effective strategies.
Collapse
Affiliation(s)
- Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, and the Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC, Australia
| | - Kylie M Quinn
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, and the Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC, Australia.
| |
Collapse
|
33
|
Ueno H. Tfh cell response in influenza vaccines in humans: what is visible and what is invisible. Curr Opin Immunol 2019; 59:9-14. [PMID: 30921542 DOI: 10.1016/j.coi.2019.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/11/2019] [Accepted: 02/22/2019] [Indexed: 01/03/2023]
Abstract
Elucidating the immune mechanism by which seasonal influenza vaccines induce a protective immune response is of great importance to gain insights into the design of next-generation vaccines conferring more effective and long-lasting immune protection. Recent studies have established that T follicular helper (Tfh) cells play a major role for the generation of antibody response following influenza vaccination. Yet, the evidence is gained largely through the analysis of blood samples, and our knowledge on the role of Tfh cells in influenza vaccination is still largely limited to the generation of antigen-specific plasmablasts. Recently, influenza vaccination was shown to induce the expansion of two types of memory B cells in addition to plasmablasts. It is plausible that activated Tfh cells that remain in the lymph nodes after vaccination, a cell population missed in the analysis of blood samples, might also contribute to the diversification of memory B cell repertoire. However, current evidence shows no increase of somatic hypermutation of the expanded memory B cell clones, suggesting that this mechanism is not efficiently active in current influenza vaccines.
Collapse
Affiliation(s)
- Hideki Ueno
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Box: 1124, Annenberg Building, Room: 15-14A New York, NY 10029, United States; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, Box: 1124, Annenberg Building, Room: 15-14A New York, NY 10029, United States.
| |
Collapse
|
34
|
Parvandeh S, Poland GA, Kennedy RB, McKinney BA. Multi-Level Model to Predict Antibody Response to Influenza Vaccine Using Gene Expression Interaction Network Feature Selection. Microorganisms 2019; 7:microorganisms7030079. [PMID: 30875727 PMCID: PMC6462975 DOI: 10.3390/microorganisms7030079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/24/2019] [Accepted: 03/08/2019] [Indexed: 11/18/2022] Open
Abstract
Vaccination is an effective prevention of influenza infection. However, certain individuals develop a lower antibody response after vaccination, which may lead to susceptibility to subsequent infection. An important challenge in human health is to find baseline gene signatures to help identify individuals who are at higher risk for infection despite influenza vaccination. We developed a multi-level machine learning strategy to build a predictive model of vaccine response using pre−vaccination antibody titers and network interactions between pre−vaccination gene expression levels. The first-level baseline−antibody model explains a significant amount of variation in post-vaccination response, especially for subjects with large pre−existing antibody titers. In the second level, we clustered individuals based on pre−vaccination antibody titers to focus gene−based modeling on individuals with lower baseline HAI where additional response variation may be predicted by baseline gene expression levels. In the third level, we used a gene−association interaction network (GAIN) feature selection algorithm to find the best pairs of genes that interact to influence antibody response within each baseline titer cluster. We used ratios of the top interacting genes as predictors to stabilize machine learning model generalizability. We trained and tested the multi-level approach on data with young and older individuals immunized against influenza vaccine in multiple cohorts. Our results indicate that the GAIN feature selection approach improves model generalizability and identifies genes enriched for immunologically relevant pathways, including B Cell Receptor signaling and antigen processing. Using a multi-level approach, starting with a baseline HAI model and stratifying on baseline HAI, allows for more targeted gene−based modeling. We provide an interactive tool that may be extended to other vaccine studies.
Collapse
Affiliation(s)
- Saeid Parvandeh
- Tandy School of Computer Science, University of Tulsa, Tulsa, OK 74104, USA.
| | - Greg A Poland
- Mayo Vaccine Group, Mayo Clinic, Rochester, MN 55905, USA.
| | | | - Brett A McKinney
- Tandy School of Computer Science, University of Tulsa, Tulsa, OK 74104, USA.
- Department of Mathematics, University of Tulsa, Tulsa, OK 74104, USA.
| |
Collapse
|
35
|
Sánchez-Vargas LA, Mathew A. Peripheral follicular helper T cells in acute viral diseases: a perspective on dengue. Future Virol 2019; 14:161-169. [PMID: 31073324 DOI: 10.2217/fvl-2018-0197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/06/2019] [Indexed: 11/21/2022]
Abstract
Follicular helper T cells (TFH) are a predominant subset of CD4+ T cells specialized in providing help to B cells in germinal centers and necessary to generate T cell-dependent antibody responses. Peripheral TFH (pTFH) are the counterpart of TFH found in the circulation, which resemble TFH in many aspects of their phenotype and function. The CD4+ pTFH subset has received a lot of interest recently because they are easy to access and have the potential to serve as a biomarker for long-lasting humoral immunity. This review will discuss recent findings of pTFH in human acute viral diseases with a focus on dengue infection.
Collapse
Affiliation(s)
- Luis A Sánchez-Vargas
- Department of Cell & Molecular Biology, Institute for Immunology & Informatics, University of Rhode Island, Providence, RI 02903, USA
| | - Anuja Mathew
- Department of Cell & Molecular Biology, Institute for Immunology & Informatics, University of Rhode Island, Providence, RI 02903, USA
| |
Collapse
|
36
|
Samson SI, Leventhal PS, Salamand C, Meng Y, Seet BT, Landolfi V, Greenberg D, Hollingsworth R. Immunogenicity of high-dose trivalent inactivated influenza vaccine: a systematic review and meta-analysis. Expert Rev Vaccines 2019; 18:295-308. [PMID: 30689467 DOI: 10.1080/14760584.2019.1575734] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/25/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION High-dose trivalent, inactivated, split-virus influenza vaccine (IIV3-HD) has been available in the US since 2009 for adults aged ≥ 65 years. To better understand how IIV3-HD provides improved protection against influenza, we systematically reviewed clinical studies comparing immune responses to IIV3-HD and standard-dose trivalent vaccine (IIV3-SD). AREAS COVERED The primary objective was to determine the relative hemagglutination inhibition (HAI) antibody response of IIV3-HD vs. IIV3-SD in adults aged ≥ 65 years. Based on seven randomized studies including more than 18,500 adults aged ≥ 65 years, combined HAI geometric mean titer (GMT) ratios (95% confidence interval) approximately 1 month post-vaccination were 1.74 (1.65-1.83) for influenza A/H1N1, 1.84 (1.73-1.95) for influenza A/H3N2, and 1.47 (1.36-1.58) for influenza B. HAI GMT ratios in these studies were similar irrespective of sex, older age (≥ 75 years), frailty, and underlying conditions. Trends were similar for A/H3N2 neutralization and anti-neuraminidase antibody titers. In immunocompromised individuals, HAI GMT ratios were mostly > 1. EXPERT OPINION In agreement with its improved efficacy and effectiveness, IIV3-HD is consistently more immunogenic than IIV3-SD in adults aged ≥ 65 years. IIV3-HD also appears more immunogenic in immunocompromised individuals.
Collapse
Affiliation(s)
| | | | | | - Ya Meng
- a Sanofi Pasteur , Swiftwater , PA , USA
| | - Bruce T Seet
- a Sanofi Pasteur , Swiftwater , PA , USA
- d Department of Molecular Genetics , University of Toronto, Medical Science Building , Toronto , ON , Canada
| | | | - David Greenberg
- a Sanofi Pasteur , Swiftwater , PA , USA
- e Department of Pediatrics , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | | |
Collapse
|
37
|
mRNA as a Transformative Technology for Vaccine Development to Control Infectious Diseases. Mol Ther 2019; 27:757-772. [PMID: 30803823 DOI: 10.1016/j.ymthe.2019.01.020] [Citation(s) in RCA: 307] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
In the last two decades, there has been growing interest in mRNA-based technology for the development of prophylactic vaccines against infectious diseases. Technological advancements in RNA biology, chemistry, stability, and delivery systems have accelerated the development of fully synthetic mRNA vaccines. Potent, long-lasting, and safe immune responses observed in animal models, as well as encouraging data from early human clinical trials, make mRNA-based vaccination an attractive alternative to conventional vaccine approaches. Thanks to these data, together with the potential for generic, low-cost manufacturing processes and the completely synthetic nature, the prospects for mRNA vaccines are very promising. In addition, mRNA vaccines have the potential to streamline vaccine discovery and development, and facilitate a rapid response to emerging infectious diseases. In this review, we overview the unique attributes of mRNA vaccine approaches, review the data of mRNA vaccines against infectious diseases, discuss the current challenges, and highlight perspectives about the future of this promising technology.
Collapse
|
38
|
Milagres L, Silva G, Pereira-Manfro W, Frota AC, Hofer C, Ferreira B, Barreto D, Figueredo M, Coelho B, Villela L, Petrovas C, Koup R. Baseline Circulating Activated TFH and Tissue-Like Exhausted B Cells Negatively Correlate With Meningococcal C Conjugate Vaccine Induced Antibodies in HIV-Infected Individuals. Front Immunol 2018; 9:2500. [PMID: 30420858 PMCID: PMC6215828 DOI: 10.3389/fimmu.2018.02500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/10/2018] [Indexed: 01/03/2023] Open
Abstract
Since 2006, meningococcal serogroup C (MenC) conjugate (MCC) vaccines have been supplied by the Brazilian government for HIV-infected children under 13 years old. For measuring protection against MenC, the serum bactericidal antibody (SBA) assay is the method of choice. The characterization of T follicular helper cells (TFH) cells has been an area of intensive study because of their significance in multiple human diseases and in vaccinology. The objective of this study was to characterize the phenotype of peripheral TFH cells and B cells and how they associated with each other and with SBA levels induced by vaccination as well as with serum cytokine levels of HIV-infected and non-infected children and adolescents. We found that CD27−IgD−CD21−CD38+ (exhausted B cells) as well as short-lived plasmablasts (CD27+IgD−CD21−CD38+) are increased in cART treated HIV patients and negatively associated with MCC vaccine induced SBA levels. Baseline frequency of activated peripheral TFH cells was a negative correlate for SBA response to MCC vaccine but positively correlated with circulating plasmablast frequency. Baseline IL4-levels positively associated with SBA response but showed a negative correlation with activated peripheral TFH cells frequency. The increased frequency of activated peripheral TFH cells found in non-responders to the vaccine implies that higher activation/differentiation of CD4 T cells within the lymph node is not necessarily associated with induction of vaccine responses.
Collapse
Affiliation(s)
- Lucimar Milagres
- Department of Microbiology Immunology and Parasitology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.,Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| | - Giselle Silva
- Department of Microbiology Immunology and Parasitology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wânia Pereira-Manfro
- Department of Microbiology Immunology and Parasitology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Cristina Frota
- Department of Pediatrics, Instituto de Puericultura e Pediatria Martagão Gesteira, Rio de Janeiro, Brazil
| | - Cristina Hofer
- Department of Pediatrics, Instituto de Puericultura e Pediatria Martagão Gesteira, Rio de Janeiro, Brazil.,Preventive Medicine Department, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bianca Ferreira
- Department of Pediatrics, Instituto de Puericultura e Pediatria Martagão Gesteira, Rio de Janeiro, Brazil
| | - Daniela Barreto
- Department of Pediatrics, Instituto de Puericultura e Pediatria Martagão Gesteira, Rio de Janeiro, Brazil
| | - Marcelo Figueredo
- Department of Periodontics, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Barbara Coelho
- Department of Periodontics, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucia Villela
- Laboratory of Immunology, Pedro Ernesto University Hospital, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Constantinos Petrovas
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| | - Richard Koup
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, United States
| |
Collapse
|
39
|
Kwun J, Park J, Yi JS, Farris AB, Kirk AD, Knechtle SJ. IL-21 Biased Alemtuzumab Induced Chronic Antibody-Mediated Rejection Is Reversed by LFA-1 Costimulation Blockade. Front Immunol 2018; 9:2323. [PMID: 30374350 PMCID: PMC6196291 DOI: 10.3389/fimmu.2018.02323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/18/2018] [Indexed: 11/25/2022] Open
Abstract
Despite its excellent efficacy in controlling T cell mediated acute rejection, lymphocyte depletion may promote a humoral response. While T cell repopulation after depletion has been evaluated in many aspects, the B cell response has not been fully elucidated. We tested the hypothesis that the mechanisms also involve skewed T helper phenotype after lymphocytic depletion. Post-transplant immune response was measured from alemtuzumab treated hCD52Tg cardiac allograft recipients with or without anti-LFA-1 mAb. Alemtuzumab induction promoted serum DSA, allo-B cells, and CAV in humanized CD52 transgenic (hCD52Tg) mice after heterotopic heart transplantation. Additional anti-LFA-1 mAb treatment resulted in reduced DSA (Fold increase 4.75 ± 6.9 vs. 0.7 ± 0.5; p < 0.01), allo-specific B cells (0.07 ± 0.06 vs. 0.006 ± 0.002 %; p < 0.01), neo-intimal hyperplasia (56 ± 14% vs. 23 ± 13%; p < 0.05), arterial disease (77.8 ± 14.2 vs. 25.8 ± 20.1%; p < 0.05), and fibrosis (15 ± 23.3 vs. 4.3 ± 1.65%; p < 0.05) in this alemtuzumab-induced chronic antibody-mediated rejection (CAMR) model. Surprisingly, elevated serum IL-21 levels in alemtuzumab-treated mice was reduced with LFA-1 blockade. In accordance with the increased serum IL-21 level, alemtuzumab treated mice showed hyperplastic germinal center (GC) development, while the supplemental anti-LFA-1 mAb significantly reduced the GC frequency and size. We report that the incomplete T cell depletion inside of the GC leads to a systemic IL-21 dominant milieu with hyperplastic GC formation and CAMR. Conventional immunosuppression, such as tacrolimus and rapamycin, failed to reverse AMR, while co-stimulation blockade with LFA-1 corrected the GC hyperplastic response. The identification of IL-21 driven chronic AMR elucidates a novel mechanism that suggests a therapeutic approach with cytolytic induction.
Collapse
Affiliation(s)
- Jean Kwun
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Jaeberm Park
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - John S Yi
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Alton B Farris
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Allan D Kirk
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| | - Stuart J Knechtle
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
40
|
T follicular helper cell development and functionality in immune ageing. Clin Sci (Lond) 2018; 132:1925-1935. [PMID: 30185614 DOI: 10.1042/cs20171157] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/24/2018] [Accepted: 08/01/2018] [Indexed: 12/15/2022]
Abstract
By 2050, there will be over 1.6 billion adults aged 65 years and older, making age-related diseases and conditions a growing public health concern. One of the leading causes of death in the ageing population is pathogenic infections (e.g. influenza, Streptococcus pneumoniae). This age-dependent susceptibility to infection has been linked to a reduced ability of the ageing immune system to mount protective responses against infectious pathogens, as well as to vaccines against these pathogens. The primary immune response that promotes protection is the production of antibodies by B cells - a response that is directly mediated by T follicular helper (TFH) cells within germinal centers (GCs) in secondary lymphoid tissues. In this review, we will summarize the current knowledge on the development and functionality of TFH cells, the use of circulating TFH (cTFH) cells as vaccine biomarkers, and the influence of age on these processes. Moreover, we will discuss the strategies for overcoming TFH cell dysfunction to improve protective antibody responses in the ageing human population.
Collapse
|
41
|
Sant AJ, Richards KA, Nayak J. Distinct and complementary roles of CD4 T cells in protective immunity to influenza virus. Curr Opin Immunol 2018; 53:13-21. [PMID: 29621639 PMCID: PMC6141328 DOI: 10.1016/j.coi.2018.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/17/2018] [Accepted: 03/19/2018] [Indexed: 02/01/2023]
Abstract
CD4 T cells play a multiplicity of roles in protective immunity to influenza. Included in these functions are help for high affinity antibody production, enhancement of CD8 T cell expansion, function and memory, acceleration of the early innate response to infection and direct cytotoxicity. The influenza-specific CD4 T cell repertoire in humans established through exposures to infection and vaccination has been found to be highly variable in abundance, specificity and functionality. Deficits in particular subsets of CD4 T cells recruited into the response result in diminished antibody responses and protection from infection. Therefore, improved strategies for vaccination should include better methods to identify deficiencies in the circulating CD4 T cell repertoire, and vaccine constructs that increase the representation of CD4 T cells of the correct specificity and functionality.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA.
| | - Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA
| | - Jennifer Nayak
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA; Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, USA
| |
Collapse
|
42
|
Moysi E, Pallikkuth S, De Armas LR, Gonzalez LE, Ambrozak D, George V, Huddleston D, Pahwa R, Koup RA, Petrovas C, Pahwa S. Altered immune cell follicular dynamics in HIV infection following influenza vaccination. J Clin Invest 2018; 128:3171-3185. [PMID: 29911996 PMCID: PMC6025971 DOI: 10.1172/jci99884] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/25/2018] [Indexed: 12/29/2022] Open
Abstract
HIV infection changes the lymph node (LN) tissue architecture, potentially impairing the immunologic response to antigenic challenge. The tissue-resident immune cell dynamics in virologically suppressed HIV+ patients on combination antiretroviral therapy (cART) are not clear. We obtained LN biopsies before and 10 to 14 days after trivalent seasonal influenza immunization from healthy controls (HCs) and HIV+ volunteers on cART to investigate CD4+ T follicular helper (Tfh) and B cell dynamics by flow cytometry and quantitative imaging analysis. Prior to vaccination, compared with those in HCs, HIV+ LNs exhibited an altered follicular architecture, but harbored higher numbers of Tfh cells and increased IgG+ follicular memory B cells. Moreover, Tfh cell numbers were dependent upon preservation of the follicular dendritic cell (FDC) network and were predictive of the magnitude of the vaccine-induced IgG responses. Interestingly, postvaccination LN samples in HIV+ participants had significantly (P = 0.0179) reduced Tfh cell numbers compared with prevaccination samples, without evidence for peripheral Tfh (pTfh) cell reduction. We conclude that influenza vaccination alters the cellularity of draining LNs of HIV+ persons in conjunction with development of antigen-specific humoral responses. The underlying mechanism of Tfh cell decline warrants further investigation, as it could bear implications for the rational design of HIV vaccines.
Collapse
Affiliation(s)
- Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Lesley R. De Armas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Louis E. Gonzalez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - David Ambrozak
- Immunology Laboratory, VRC, NIAID, NIH, Bethesda, Maryland, USA
| | - Varghese George
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - David Huddleston
- Department of Trauma Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rajendra Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Richard A. Koup
- Immunology Laboratory, VRC, NIAID, NIH, Bethesda, Maryland, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
43
|
Harnessing T Follicular Helper Cell Responses for HIV Vaccine Development. Viruses 2018; 10:v10060336. [PMID: 29921828 PMCID: PMC6024737 DOI: 10.3390/v10060336] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 12/12/2022] Open
Abstract
Passive administration of broadly neutralizing antibodies (bNAbs) capable of recognizing a broad range of viral strains to non-human primates has led to protection from infection with chimeric SIV/HIV virus (SHIV). This data suggests that generating protective antibody responses could be an effective strategy for an HIV vaccine. However, classic vaccine approaches have failed so far to induce such protective antibodies in HIV vaccine trials. HIV-specific bNAbs identified in natural infection show high levels of somatic hypermutations, demonstrating that they underwent extensive affinity maturation. It is likely that to gain ability to recognize diverse viral strains, vaccine-induced humoral responses will also require complex, iterative maturation. T follicular helper cells (Tfh) are a specialized CD4+ T cell subset that provides help to B cells in the germinal center for the generation of high-affinity and long-lasting humoral responses. It is therefore probable that the quality and quantity of Tfh responses upon vaccination will impact development of bNAbs. Here, we review studies that advanced our understanding of Tfh differentiation, function and regulation. We discuss correlates of Tfh responses and bNAb development in natural HIV infection. Finally, we highlight recent strategies to optimize Tfh responses upon vaccination and their impact on prophylactic HIV vaccine research.
Collapse
|
44
|
Cárdeno A, Magnusson MK, Quiding-Järbrink M, Lundgren A. Activated T follicular helper-like cells are released into blood after oral vaccination and correlate with vaccine specific mucosal B-cell memory. Sci Rep 2018; 8:2729. [PMID: 29426881 PMCID: PMC5807513 DOI: 10.1038/s41598-018-20740-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
T follicular helper (Tfh)-like cells with potent B-cell helping ability are mobilized into human circulation after parenteral vaccination and are generally held to reflect ongoing germinal center reactions. However, whether mucosal vaccination induces systemic Tfh responses and how such responses may relate to IgA production are unknown. We investigated the frequencies, phenotype and function of circulating Tfh-like CD4+CXCR5+ T cells (cTfh) in adults receiving an oral inactivated enterotoxigenic Escherichia coli vaccine. Subjects were classified as vaccine responders or weak/non-responders based on their intestine-derived antibody-secreting cell (ASC) IgA responses to major vaccine antigens. Oral immunization induced significantly increased proportions of cTfh cells expressing the cTfh activation marker inducible costimulator (ICOS) in ASC responders, but not in weak/non-responders. Vaccination also enhanced the expression of IL-21, Th17 markers and integrin β7 by activated cTfh cells, supporting functionality and gut homing potential. cTfh cells promoted total and vaccine specific IgA production from cocultured B cells. Magnitudes of cTfh responses assessed within a week after primary vaccinations correlated with memory intestine-derived vaccine specific IgA responses 1-2 years later. We conclude that activated ICOS+ Tfh-like cells are mobilized into blood after oral vaccination and may be used as biomarkers of vaccine specific mucosal memory in humans.
Collapse
Affiliation(s)
- Ana Cárdeno
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Maria K Magnusson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | | | - Anna Lundgren
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
45
|
Ylipalosaari P, Ala-Kokko TI, Laurila J, Ahvenjärvi L, Syrjälä H. ICU-treated influenza A(H1N1) pdm09 infections more severe post pandemic than during 2009 pandemic: a retrospective analysis. BMC Infect Dis 2017; 17:728. [PMID: 29162037 PMCID: PMC5697104 DOI: 10.1186/s12879-017-2829-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/12/2017] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND We compared in a single mixed intensive care unit (ICU) patients with influenza A(H1N1) pdm09 between pandemic and postpandemic periods. METHODS Retrospective analysis of prospectively collected data in 2009-2016. Data are expressed as median (25th-75th percentile) or number (percentile). RESULTS Seventy-six influenza A(H1N1) pdm09 patients were admitted to the ICU: 16 during the pandemic period and 60 during the postpandemic period. Postpandemic patients were significantly older (60 years vs. 43 years, p < 0.001) and less likely to have epilepsy or other neurological diseases compared with pandemic patients (5 [8.3%] vs. 6 [38%], respectively; p = 0.009). Postpandemic patients were more likely than pandemic patients to have cardiovascular disease (24 [40%] vs. 1 [6%], respectively; p = 0.015), and they had higher scores on APACHE II (17 [13-22] vs. 14 [10-17], p = 0.002) and SAPS II (40 [31-51] vs. 31 [25-35], p = 0.002) upon admission to the ICU. Postpandemic patients had higher maximal SOFA score (9 [5-12] vs. 5 [4-9], respectively; p = 0.03) during their ICU stay. Postpandemic patients had more often septic shock (40 [66.7%] vs. 8 [50.0%], p = 0.042), and longer median hospital stays (15.0 vs. 8.0 days, respectively; p = 0.006). During 2015-2016, only 18% of the ICU- treated patients had received seasonal influenza vaccination. CONCLUSIONS Postpandemic ICU-treated A(H1N1) pdm09 influenza patients were older and developed more often septic shock and had longer hospital stays than influenza patients during the 2009 pandemic.
Collapse
Affiliation(s)
- Pekka Ylipalosaari
- Department of Infection Control, Oulu University Hospital, Box 21, FIN-90029 Oulu, OYS Finland
| | - Tero I. Ala-Kokko
- Department of Anesthesiology, Division of Intensive Care, Oulu University Hospital, FIN-90029 Oulu, OYS Finland
- Medical Research Center, Research Group of Surgery, Anesthesiology and Intensive Care, University of Oulu, Oulu, Finland
| | - Jouko Laurila
- Department of Anesthesiology, Division of Intensive Care, Oulu University Hospital, FIN-90029 Oulu, OYS Finland
- Medical Research Center, Research Group of Surgery, Anesthesiology and Intensive Care, University of Oulu, Oulu, Finland
| | - Lauri Ahvenjärvi
- Department of Radiology, Oulu University Hospital, FIN-90029 Oulu, OYS Finland
| | - Hannu Syrjälä
- Department of Infection Control, Oulu University Hospital, Box 21, FIN-90029 Oulu, OYS Finland
| |
Collapse
|
46
|
Lindgren G, Ols S, Liang F, Thompson EA, Lin A, Hellgren F, Bahl K, John S, Yuzhakov O, Hassett KJ, Brito LA, Salter H, Ciaramella G, Loré K. Induction of Robust B Cell Responses after Influenza mRNA Vaccination Is Accompanied by Circulating Hemagglutinin-Specific ICOS+ PD-1+ CXCR3+ T Follicular Helper Cells. Front Immunol 2017; 8:1539. [PMID: 29181005 PMCID: PMC5693886 DOI: 10.3389/fimmu.2017.01539] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 10/27/2017] [Indexed: 12/22/2022] Open
Abstract
Modified mRNA vaccines have developed into an effective and well-tolerated vaccine platform that offers scalable and precise antigen production. Nevertheless, the immunological events leading to strong antibody responses elicited by mRNA vaccines are largely unknown. In this study, we demonstrate that protective levels of antibodies to hemagglutinin were induced after two immunizations of modified non-replicating mRNA encoding influenza H10 encapsulated in lipid nanoparticles (LNP) in non-human primates. While both intradermal (ID) and intramuscular (IM) administration induced protective titers, ID delivery generated this response more rapidly. Circulating H10-specific memory B cells expanded after each immunization, along with a transient appearance of plasmablasts. The memory B cell pool waned over time but remained detectable throughout the 25-week study. Following prime immunization, H10-specific plasma cells were found in the bone marrow and persisted over time. Germinal centers were formed in vaccine-draining lymph nodes along with an increase in circulating H10-specific ICOS+ PD-1+ CXCR3+ T follicular helper cells, a population shown to correlate with high avidity antibody responses after seasonal influenza vaccination in humans. Collectively, this study demonstrates that mRNA/LNP vaccines potently induce an immunological repertoire associated with the generation of high magnitude and quality antibodies.
Collapse
Affiliation(s)
- Gustaf Lindgren
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Frank Liang
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth A Thompson
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ang Lin
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrika Hellgren
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kapil Bahl
- Valera LLC, Cambridge, MA, United States
| | - Shinu John
- Valera LLC, Cambridge, MA, United States
| | | | | | - Luis A Brito
- Moderna Therapeutics, Cambridge, MA, United States
| | - Hugh Salter
- Moderna Therapeutics, Cambridge, MA, United States.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Karin Loré
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Merani S, Kuchel GA, Kleppinger A, McElhaney JE. Influenza vaccine-mediated protection in older adults: Impact of influenza infection, cytomegalovirus serostatus and vaccine dosage. Exp Gerontol 2017; 107:116-125. [PMID: 28958701 DOI: 10.1016/j.exger.2017.09.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/22/2017] [Accepted: 09/24/2017] [Indexed: 12/19/2022]
Abstract
Age-related changes in T-cell function are associated with a loss of influenza vaccine efficacy in older adults. Both antibody and cell-mediated immunity plays a prominent role in protecting older adults, particularly against the serious complications of influenza. High dose (HD) influenza vaccines induce higher antibody titers in older adults compared to standard dose (SD) vaccines, yet its impact on T-cell memory is not clear. The aim of this study was to compare the antibody and T-cell responses in older adults randomized to receive HD or SD influenza vaccine as well as determine whether cytomegalovirus (CMV) serostatus affects the response to vaccination, and identify differences in the response to vaccination in those older adults who subsequently have an influenza infection. Older adults (≥65years) were enrolled (n=106) and randomized to receive SD or HD influenza vaccine. Blood was collected pre-vaccination, followed by 4, 10 and 20weeks post-vaccination. Serum antibody titers, as well as levels of inducible granzyme B (iGrB) and cytokines were measured in PBMCs challenged ex vivo with live influenza virus. Surveillance conducted during the influenza season identified those with laboratory confirmed influenza illness or infection. HD influenza vaccination induced a high antibody titer and IL-10 response, and a short-lived increase in Th1 responses (IFN-γ and iGrB) compared to SD vaccination in PBMCs challenged ex vivo with live influenza virus. Of the older adults who became infected with influenza, a high IL-10 and iGrB response in virus-challenged cells was observed post-infection (week 10 to 20), as well as IFN-γ and TNF-α at week 20. Additionally, CMV seropositive older adults had an impaired iGrB response to influenza virus-challenge, regardless of vaccine dose. This study illustrates that HD influenza vaccines have little impact on the development of functional T-cell memory in older adults. Furthermore, poor outcomes of influenza infection in older adults may be due to a strong IL-10 response to influenza following vaccination, and persistent CMV infection.
Collapse
Affiliation(s)
- Shahzma Merani
- Health Sciences North Research Institute, 41 Ramsey Lake Road, Sudbury, P3E 5J1, ON, Canada
| | - George A Kuchel
- University of Connecticut Center on Aging, UConn Health, 263 Farmington Avenue, Farmington, 06030-5215, CT, USA
| | | | - Janet E McElhaney
- Health Sciences North Research Institute, 41 Ramsey Lake Road, Sudbury, P3E 5J1, ON, Canada.
| |
Collapse
|
48
|
Abstract
T follicular helper (Tfh) cells are a distinct type of CD4+ T cell specialized in providing help to B cells during the germinal centre (GC) reaction. As such, they are critical determinants of the quality of an antibody response following antigen challenge. Excessive production of Tfh cells can result in autoimmunity whereas too few can result in inadequate protection from infection. Hence, their differentiation and maintenance must be tightly regulated to ensure appropriate but limited help to B cells. Unlike the majority of other CD4+ T-cell subsets, Tfh cell differentiation occurs in three phases defined by their anatomical location. During each phase of differentiation the emerging Tfh cells express distinct patterns of co-receptors, which work together with the T-cell receptor (TCR) to drive Tfh differentiation. These signals provided by both TCR and co-receptors during Tfh differentiation alter proliferation, survival, metabolism, cytokine production and transcription factor expression. This review will discuss how engagement of TCR and co-receptors work together to shape the formation and function of Tfh cells.
Collapse
Affiliation(s)
- Louise M C Webb
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Michelle A Linterman
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| |
Collapse
|
49
|
Merani S, Pawelec G, Kuchel GA, McElhaney JE. Impact of Aging and Cytomegalovirus on Immunological Response to Influenza Vaccination and Infection. Front Immunol 2017; 8:784. [PMID: 28769922 PMCID: PMC5512344 DOI: 10.3389/fimmu.2017.00784] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 06/21/2017] [Indexed: 12/13/2022] Open
Abstract
The number of people over the age of 60 is expected to double by 2050 according to the WHO. This emphasizes the need to ensure optimized resilience to health stressors in late life. In older adults, influenza is one of the leading causes of catastrophic disability (defined as the loss of independence in daily living and self-care activities). Influenza vaccination is generally perceived to be less protective in older adults, with some studies suggesting that the humoral immune response to the vaccine is further impaired in cytomegalovirus (CMV)-seropositive older people. CMV is a β-herpes virus infection that is generally asymptomatic in healthy individuals. The majority of older adults possess serum antibodies against the virus indicating latent infection. Age-related changes in T-cell-mediated immunity are augmented by CMV infection and may be associated with more serious complications of influenza infection. This review focuses on the impact of aging and CMV on immune cell function, the response to influenza infection and vaccination, and how the current understanding of aging and CMV can be used to design a more effective influenza vaccine for older adults. It is anticipated that efforts in this field will address the public health need for improved protection against influenza in older adults, particularly with regard to the serious complications leading to loss of independence.
Collapse
Affiliation(s)
- Shahzma Merani
- Health Sciences North Research Institute, Sudbury, ON, Canada
| | - Graham Pawelec
- Health Sciences North Research Institute, Sudbury, ON, Canada.,Second Department of Internal Medicine, University of Tübingen Medical Center, Tübingen, Germany
| | - George A Kuchel
- UConn Center on Aging, UConn Health, Farmington, CT, United States
| | | |
Collapse
|