1
|
Matz HC, Ellebedy AH. Vaccination against influenza viruses annually: Renewing or narrowing the protective shield? J Exp Med 2025; 222:e20241283. [PMID: 40272481 PMCID: PMC12020744 DOI: 10.1084/jem.20241283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Annual vaccines are recommended for the seasonal influenza virus. While yearly updates to the vaccine are necessary due to the constant evolution of influenza viruses, some studies have suggested repeat vaccination may result in a reduction in vaccine effectiveness in subsequent years. This review examines the available evidence that repeated annual influenza virus vaccination may have effects on future vaccine responses, and it synthesizes the available data with studies that may indicate potential immunological mechanisms underlying these effects. The goal is to examine the available literature to determine whether these mechanisms can be subverted to improve seasonal influenza virus vaccine efficacy.
Collapse
Affiliation(s)
- Hanover C. Matz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
2
|
Zasztowt-Sternicka M, Jagielska A, Rząd M, Szymusik I, Hallmann E, Brydak L, Nitsch-Osuch A. Immunogenicity of inactivated quadrivalent influenza vaccine in pregnant women, including the level of postvaccination antibodies in umbilical cord blood. Vaccine 2025; 53:127047. [PMID: 40203592 DOI: 10.1016/j.vaccine.2025.127047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/28/2024] [Accepted: 03/13/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Infants and pregnant women are particularly vulnerable to severe illness and complications of influenza. It is necessary to increase knowledge about the safety and effectiveness of influenza vaccination during pregnancy to improve awareness among patients and healthcare professionals and increase to increase vaccination coverage. METHODS A prospective, single-group clinical study was performed in the 2021/2022 and 2022/2023 seasons. The aim of the study was to assess the immunogenicity and tolerance of quadrivalent influenza vaccine (IIV4) in pregnant women, to evaluate transplacental antibody (AB) transfer after IIV4 administration, and to estimate optimal gestational age for vaccination. A group of 62 pregnant women between 14 and 37 weeks of gestation, including 59 mother-child pairs, underwent vaccination with IIV4. The humoral immune response was assessed using the EMA criteria. RESULTS Baseline seroprotection for all IIV4 strains was significantly correlated with previous vaccinations (p < .5). The postvaccination seroprotection ranged between 71 % and 94 %. A significant increase in AB titers was observed for all vaccine strains (p < .0000). No significant differences were observed in seroprotection and seroconversion rates or transplacental AB transfer between women vaccinated in the 2nd vs 3rd trimester of pregnancy. All IIV4 strains used in the study met at least 1 of the 3 EMA/CPMP immunogenicity criteria and may thus be considered immunogenic. Seroprotection for A/H1N1/pdm09, A/H3N2, B/lineage Victoria (B/V), and B/lineage Yamagata (B/Y) antigens was achieved by 97 %, 95 %, 59 %, and 85 % of neonates, respectively. Over 43 % of participants reported solicited injection site reactions. None of the participants reported severe or unsolicited adverse events. CONCLUSIONS IIV4 induced a strong immune response in pregnant women and a high rate of seroprotection in neonates. Immune protection in children did not differ according to the trimester of vaccination in pregnancy. IIV4 was well tolerated by pregnant women.
Collapse
Affiliation(s)
- Monika Zasztowt-Sternicka
- Doctoral School, Medical University of Warsaw, Poland; Department of Social Medicine and Public Health, Medical University of Warsaw, Poland; 1(st) Chair and Department of Obstetrics and Gynecology, Medical University of Warsaw, Poland.
| | | | - Michał Rząd
- Doctoral School, Medical University of Warsaw, Poland; Department of Social Medicine and Public Health, Medical University of Warsaw, Poland
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Ewelina Hallmann
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | - Lidia Brydak
- Department of Virology, National Institute of Public Health NIH-National Research Institute, Warsaw, Poland
| | | |
Collapse
|
3
|
Goodwin E, Gibbs JS, Yewdell JW, Eisenlohr LC, Hensley SE. Influenza virus antibodies inhibit antigen-specific de novo B cell responses in mice. J Virol 2024; 98:e0076624. [PMID: 39194245 PMCID: PMC11406888 DOI: 10.1128/jvi.00766-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Antibody responses to influenza vaccines tend to be focused on epitopes encountered during prior influenza exposures, with little production of de novo responses to novel epitopes. To examine the contribution of circulating antibodies to this phenomenon, we passively transferred a hemagglutinin (HA)-specific monoclonal antibody (mAb) into mice before immunizing with whole inactivated virions. The HA mAb inhibited de novo HA-specific antibodies, plasmablasts, germinal center B cells, and memory B cells, while responses to a second antigen in the vaccine, neuraminidase (NA), were uninhibited. The HA mAb potently inhibited de novo antibody responses against epitopes near the HA mAb binding site. The HA mAb also promoted IgG1 class switching, an effect that, unlike the inhibition of HA responses, relied on signaling through Fc-gamma receptors. These studies suggest that circulating antibodies inhibit de novo B cell responses in an antigen-specific manner, which likely contributes to differences in antibody specificities elicited during primary and secondary influenza virus exposures.IMPORTANCEMost humans are exposed to influenza viruses in childhood and then subsequently exposed to antigenically drifted influenza variants later in life. It is unclear if antibodies elicited by earlier influenza virus exposures impact immunity against new influenza virus strains. Here, we used a mouse model to investigate how an anti-hemagglutinin (HA) monoclonal antibody (mAb) affects de novo B cell and antibody responses to the protein targeted by the monoclonal antibody (HA) and a second protein not targeted by the monoclonal antibody [neuraminidase (NA)]. Collectively, our studies suggest that circulating anti-influenza virus antibodies can potently modulate the magnitude and specificity of antibody responses elicited by secondary influenza virus exposures.
Collapse
Affiliation(s)
- Eileen Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James S. Gibbs
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan W. Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Laurence C. Eisenlohr
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Cyster JG, Wilson PC. Antibody modulation of B cell responses-Incorporating positive and negative feedback. Immunity 2024; 57:1466-1481. [PMID: 38986442 PMCID: PMC11257158 DOI: 10.1016/j.immuni.2024.06.009] [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: 04/15/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
Antibodies are powerful modulators of ongoing and future B cell responses. While the concept of antibody feedback has been appreciated for over a century, the topic has seen a surge in interest due to the evidence that the broadening of antibody responses to SARS-CoV-2 after a third mRNA vaccination is a consequence of antibody feedback. Moreover, the discovery that slow antigen delivery can lead to more robust humoral immunity has put a spotlight on the capacity for early antibodies to augment B cell responses. Here, we review the mechanisms whereby antibody feedback shapes B cell responses, integrating findings in humans and in mouse models. We consider the major influence of epitope masking and the diverse actions of complement and Fc receptors and provide a framework for conceptualizing the ways antigen-specific antibodies may influence B cell responses to any form of antigen, in conditions as diverse as infectious disease, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Patrick C Wilson
- Drukier Institute for Children's Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Zhang T, Han Y, Huang W, Wei H, Zhao Y, Shu L, Guo Y, Ye B, Zhou J, Liu J. Neutralizing antibody responses against contemporary and future influenza A(H3N2) viruses in paradoxical clades elicited by repeated and single vaccinations. J Med Virol 2024; 96:e29743. [PMID: 38884419 DOI: 10.1002/jmv.29743] [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: 03/06/2024] [Revised: 05/16/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024]
Abstract
As one of the most effective measures to prevent seasonal influenza viruses, annual influenza vaccination is globally recommended. Nevertheless, evidence regarding the impact of repeated vaccination to contemporary and future influenza has been inconclusive. A total of 100 subjects singly or repeatedly immunized with influenza vaccines including 3C.2a1 or 3C.3a1 A(H3N2) during 2018-2019 and 2019-2020 influenza season were recruited. We investigated neutralization antibody by microneutralization assay using four antigenically distinct A(H3N2) viruses circulating from 2018 to 2023, and tracked the dynamics of B cell receptor (BCR) repertoire for consecutive vaccinations. We found that vaccination elicited cross-reactive antibody responses against future emerging strains. Broader neutralizing antibodies to A(H3N2) viruses and more diverse BCR repertoires were observed in the repeated vaccination. Meanwhile, a higher frequency of BCR sequences shared among the repeated-vaccinated individuals with consistently boosting antibody response was found than those with a reduced antibody response. Our findings suggest that repeated seasonal vaccination could broaden the breadth of antibody responses, which may improve vaccine protection against future emerging viruses.
Collapse
MESH Headings
- Humans
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Influenza, Human/virology
- Adult
- Cross Reactions/immunology
- Male
- Female
- Vaccination
- Middle Aged
- Young Adult
- Neutralization Tests
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/genetics
- Adolescent
Collapse
Affiliation(s)
- Ting Zhang
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Unit of Adaptive Evolution and Control of Emerging Viruses (2018RU009), Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Han
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Unit of Adaptive Evolution and Control of Emerging Viruses (2018RU009), Chinese Academy of Medical Sciences, Beijing, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Weijuan Huang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hejiang Wei
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yingze Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Unit of Adaptive Evolution and Control of Emerging Viruses (2018RU009), Chinese Academy of Medical Sciences, Beijing, China
| | - Liumei Shu
- Department of Health Care, Beijing Daxing District Hospital, Beijing, China
| | - Yaxin Guo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Unit of Adaptive Evolution and Control of Emerging Viruses (2018RU009), Chinese Academy of Medical Sciences, Beijing, China
| | - Beiwei Ye
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Unit of Adaptive Evolution and Control of Emerging Viruses (2018RU009), Chinese Academy of Medical Sciences, Beijing, China
| | - Jianfang Zhou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jun Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Unit of Adaptive Evolution and Control of Emerging Viruses (2018RU009), Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Fox A. Drift and shape-new insights into human immunity against influenza virus neuraminidase. mBio 2023; 14:e0165423. [PMID: 37933976 PMCID: PMC10746272 DOI: 10.1128/mbio.01654-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023] Open
Abstract
Influenza virus hemagglutinin mediates infection by binding sialic acids, whereas neuraminidase cleaves sialic acids to release progeny virions. Both are targets of protective antibodies, but influenza vaccine strain selection and antigen dose are based on hemagglutinin alone. Virus characterization using first infection ferret sera indicates that escape from hemagglutination inhibiting (HI) antibodies occurs more frequently and is not coordinated with escape from neuraminidase inhibiting (NI) antibodies. A key question addressed by Daulagala et al. (P. Daulagala, B. R. Mann, K. Leung, E. H. Y. Lau, et al., mBio 14:e00084-23, 2023, https://doi.org/10.1128/mbio.00084-23) is how this translates to humans who encounter multiple influenza viruses throughout life. Their cross-sectional study, using sera from a wide age range of participants and H1N1 viruses spanning 1977-2015, indicates that NI antibodies are more broadly cross-reactive than HI antibodies. Both HI and NI titers were highest against strains encountered in childhood indicating that both are shaped by priming exposures. The study further supports the development of NA-optimized vaccines.
Collapse
Affiliation(s)
- Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Larsen Rasmussen S, Kumar P, Trebbien R, Leutscher P, Rasmussen C. Influenza Vaccine Type-Dependent Antibody Response in Patients with Autoimmune Inflammatory Rheumatic Diseases. Eur J Rheumatol 2023; 10:122-129. [PMID: 37873666 PMCID: PMC10765197 DOI: 10.5152/eurjrheum.2023.23053] [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: 06/16/2023] [Accepted: 08/31/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND The study aimed to explore influenza antibody response in patients with autoimmune inflammatory rheumatoid diseases (AIIRDs) stratified by the different vaccine types applied in Denmark during the 2018-2019 influenza season. METHODS Included patients were diagnosed with rheumatoid arthritis, psoriatic arthritis, or spondyloarthritis receiving biological disease-modifying antirheumatic drugs (bDMARDs) with or without conventional synthetic disease-modifying antirheumatic drugs. Influenza vaccination status in the 2018-2019 season and vaccine type received were reviewed in the Denmark. Blood samples were drawn ≥ 14 days post vaccination, and antibody titers were determined by the hemagglutinin inhibition (HAI) assay for the serotypes A/Michigan/H1N1, A/Singapore/H3N2, and B/Colorado included in the influenza vaccines in the 2018-2019 season. An overall serotype HAI geometric mean titer (GMT) was calculated from the 3 serotype-specific HAI titers. An overall serotype HAI GMT ≥ 40 was considered protective. RESULTS Of the 205 included patients, 105 (51%) had received influenza vaccination. One-quarter of vaccinated patients achieved post-vaccination overall serotype HAI GMT ≥40. For patients vaccinated with Influvac, a significantly higher proportion had HAI titers ≥ 40 for 2 serotypes, namely, A/Michigan/H1N1 and A/Singapore/H3N2, than patients vaccinated with Vaxigrip or VaxigripTetra. The same applied to all serotypes HAI GMT, where significantly more patients who received Influvac achieved postvaccination HAI GMT≥40 versus patients who received Vaxigrip (p=0.02) or VaxigripTetra (p=0.002). The latter outcome was explored in a multivariable logistic regression analysis and remained significant when including the following variables: age, sex, treatment with methotrexate and/or prednisolone, type of influenza vaccine, time interval from vaccination to antibody measurement, and previous vaccination status. CONCLUSION Influenza antibody levels following vaccination with Influvac in bDMARD-treated patients with AIIRDs were superior to Vaxigrip and VaxigripTetra. Treatment with methotrexate (MTX) did not reduce the antibody response.
Collapse
Affiliation(s)
| | - Prabhat Kumar
- Department of Rheumatology, North Denmark Regional Hospital, Hjoerring, Denmark
| | - Ramona Trebbien
- National Influenza Center, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Leutscher
- North Denmark Regional Hospital, Centre for Clinical Research, Hjoerring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Claus Rasmussen
- North Denmark Regional Hospital, Centre for Clinical Research, Hjoerring, Denmark
- Department of Rheumatology, North Denmark Regional Hospital, Hjoerring, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
8
|
Frasca D, Romero M, Diaz A, Blomberg BB. Obesity accelerates age defects in B cells, and weight loss improves B cell function. Immun Ageing 2023; 20:35. [PMID: 37460937 PMCID: PMC10351107 DOI: 10.1186/s12979-023-00361-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND We have previously shown that obesity accelerates age-associated defects in B cell function and antibody production leading to decreased secretion of protective antibodies and increased autoimmunity. We wanted to evaluate if obese adults enrolled in a voluntary weight reduction program had higher protective and lower autoimmune antibody responses similar to those observed in lean adults. METHODS Experiments were performed using blood isolated from an established cohort of female lean adult and elderly individuals, as well as from the blood of female adults with obesity, before and after a voluntary weight reduction program in which their Body Mass Index (BMI) was reduced 10-34% in 12 months. All participants were vaccinated with the Trivalent Inactivated Influenza vaccine. Serum samples were evaluated for the presence of pro-inflammatory cytokines and adipokines, vaccine-specific antibodies and autoimmune antibodies. We evaluated the composition of the B cell pool by flow cytometry, the expression of RNA for class switch transcription factors and pro-inflammatory markers by qPCR, the in vitro secretion of pro- and anti-inflammatory cytokines and their capacity to induce pro-inflammatory T cells. RESULTS Obesity, similar to aging, induced increased serum levels of pro-inflammatory cytokines and autoimmune antibodies, while vaccine-specific antibodies were reduced. In agreement with the serum results, the B cell pool of obese adults and elderly individuals was enriched in pro-inflammatory B cell subsets and was characterized by higher expression of markers associated with cell senescence, higher levels of T-bet, the transcription factor for autoimmune antibodies and lower levels of E47, the transcription factor associated with protective responses to the influenza vaccine. B cells from obese adults and elderly individuals were also able to secrete inflammatory cytokines and support the generation of inflammatory T cells. All these pro-inflammatory characteristics of B cells from obese individuals were significantly attenuated, but not completely reversed, by weight loss. CONCLUSIONS Although the results from our small observational study show that obesity-induced dysfunctional B cell responses, similar to those occurring during aging, are ameliorated in some but not all obese individuals after weight loss, the effects of body weight loss on mechanistic pathways are largely missing and deserve further investigation.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, RMSB 3153, 1600 NW 10thAve, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, RMSB 3153, 1600 NW 10thAve, Miami, FL, 33136, USA
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, RMSB 3153, 1600 NW 10thAve, Miami, FL, 33136, USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, RMSB 3153, 1600 NW 10thAve, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
9
|
Kang M, Lin F, Jiang Z, Tan X, Lin X, Liang Z, Xiao C, Xia Y, Guan W, Yang Z, Yu G, Zanin M, Tang S, Wong S. The impact of pre-existing influenza antibodies and inflammatory status on the influenza vaccine responses in older adults. Influenza Other Respir Viruses 2023; 17:e13172. [PMID: 37457646 PMCID: PMC10339007 DOI: 10.1111/irv.13172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 07/18/2023] Open
Abstract
Age-associated immune changes and pre-existing influenza immunity are hypothesized to reduce influenza vaccine effectiveness in older adults, although the contribution of each factor is unknown. Here, we constructed influenza-specific IgG landscapes and determined baseline concentrations of cytokines typically associated with chronic inflammation in older adults (TNF-α, IL-10, IL-6, and IFN-γ) in 30 high and 29 low influenza vaccine responders (HR and LR, respectively). In a background of high H3 antibody titers, vaccine-specific H3, but not H1, antibody titers were boosted in LRs to titers comparable to HRs. Pre-vaccination concentrations of IL-10 were higher in LRs compared with HRs and inversely correlated with titers of pre-existing influenza antibodies. Baseline TNF-α concentrations were positively correlated with fold-increases in antibody titers in HRs. Our findings indicate that baseline inflammatory status is an important determinant for generating post-vaccination hemagglutinin-inhibition antibodies in older adults, and IgG responses can be boosted in the context of high pre-existing immunity.
Collapse
Affiliation(s)
- Min Kang
- School of Public HealthSouthern Medical UniversityGuangzhouP. R. China
- Guangdong Center for Disease Control and PreventionGuangzhouP. R. China
| | - Fangmei Lin
- Guangzhou Medical University, XinzaoGuangzhouP. R. China
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory DiseaseGuangzhouP.R. China
| | - Zhanpeng Jiang
- Guangzhou Medical University, XinzaoGuangzhouP. R. China
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory DiseaseGuangzhouP.R. China
| | - Xiaohua Tan
- Guangdong Center for Disease Control and PreventionGuangzhouP. R. China
| | - Xia Lin
- Guangzhou Medical University, XinzaoGuangzhouP. R. China
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory DiseaseGuangzhouP.R. China
| | - Zaolan Liang
- Guangzhou Medical University, XinzaoGuangzhouP. R. China
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory DiseaseGuangzhouP.R. China
- HKU‐Pasteur Research Pole, School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Cheng Xiao
- Guangzhou Medical University, XinzaoGuangzhouP. R. China
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory DiseaseGuangzhouP.R. China
- HKU‐Pasteur Research Pole, School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| | - Yonghe Xia
- Zhongshan Yiyan Bio‐Pharmaceutical Co., LtdZhongshanP. R. China
| | - Wenda Guan
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory DiseaseGuangzhouP.R. China
- Guangzhou Institute of Respiratory HealthFirst Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory MedicineGuangzhouP.R. China
| | - Zifeng Yang
- State Key Laboratory for Respiratory Diseases and National Clinical Research Centre for Respiratory DiseaseGuangzhouP.R. China
- Guangzhou Institute of Respiratory HealthFirst Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory MedicineGuangzhouP.R. China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouP. R. China
| | - Mark Zanin
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
- Centre for Immunology & InfectionShatinHong Kong
| | - Shixing Tang
- School of Public HealthSouthern Medical UniversityGuangzhouP. R. China
| | - Sook‐San Wong
- HKU‐Pasteur Research Pole, School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
- School of Public Health, LKS Faculty of MedicineThe University of Hong KongHong KongChina
| |
Collapse
|
10
|
Xu Q, Wei H, Wen S, Chen J, Lei Y, Cheng Y, Huang W, Wang D, Shu Y. Factors affecting the immunogenicity of influenza vaccines in human. BMC Infect Dis 2023; 23:211. [PMID: 37024804 PMCID: PMC10078025 DOI: 10.1186/s12879-023-08158-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND The influenza viruses pose a threat to human health and medical services, and vaccination is an important way to prevent infection. However, the effectiveness of influenza vaccines is affected by various aspects. This study aimed to explore factors related to the immune response to influenza vaccines. METHODS The study was conducted from September 2019 to September 2021, and a total of 593 volunteers were recruited from the Center for Disease Control and Prevention in 3 provinces in China. The hemagglutination inhibition assay was used to measure antibody levels. The Chi-square test, multivariable logistic regression analysis, and sum-rank test were used to analyze the factors associated with influenza vaccine immune response. RESULTS The Chi-square test showed that seroconversion rates and response rate were associated with age group, vaccination history, chronic conditions, the frequency of colds, and region (P < 0.05). The multivariable logistic regression analysis showed that age was an important factor that affected participants' seroconversion rates for A/H1N1, A/H3N2, B/Victoria, and response status (18-64 vs. ≤5: OR = 2.77, P < 0.001; ≥65 vs. ≤5: OR = 0.38, P = 0.01; 18-64 vs. ≤5: OR = 2.64, P = 0.03). Vaccination history was also an affecting factor for A/H1N1, B/Victoria, and response status (yes vs. no: OR = 0.4 / 0.44 / 0.25, P < 0.001). The frequency of colds and chronic conditions were also affecting factors for participants' seroconversion rates and response levels to different degrees. The sum-rank test showed that the fold changes for A/H1N1, B/Victoria, and B/Yamagata were associated with age group and vaccination history (P < 0.01). The fold changes for A/H3N2 were associated with the frequency of colds (P < 0.05), and those for B/Victoria were associated with gender and chronic conditions (P < 0.05). CONCLUSIONS Vaccination history, age, health condition, and frequency of colds were important factors affecting the seroconversion rate of the influenza vaccine in human. There is a need for developing optimized vaccination strategies for vulnerable groups to improve the efficacy of influenza vaccines in human.
Collapse
Affiliation(s)
- Qiuyi Xu
- School of Public Health (Shenzhen), Sun Yat-sen University-Shenzhen Campus, Shenzhen, Guangdong, China
| | - Hejiang Wei
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Simin Wen
- Clinical Research Center, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China, University of Technology, Guangzhou, Guangdong, China
| | - Jiamin Chen
- School of Public Health (Shenzhen), Sun Yat-sen University-Shenzhen Campus, Shenzhen, Guangdong, China
| | - Yuxuan Lei
- School of Public Health (Shenzhen), Sun Yat-sen University-Shenzhen Campus, Shenzhen, Guangdong, China
| | - Yanhui Cheng
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Weijuan Huang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dayan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University-Shenzhen Campus, Shenzhen, Guangdong, China.
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical School, Beijing, China.
| |
Collapse
|
11
|
Dangi T, Sanchez S, Lew MH, Awakoaiye B, Visvabharathy L, Richner JM, Koralnik IJ, Penaloza-MacMaster P. Pre-existing immunity modulates responses to mRNA boosters. Cell Rep 2023; 42:112167. [PMID: 36857186 PMCID: PMC9928730 DOI: 10.1016/j.celrep.2023.112167] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 12/19/2022] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
mRNA vaccines are effective in preventing severe COVID-19, but breakthrough infections, emerging variants, and waning immunity warrant the use of boosters. Although mRNA boosters are being implemented, the extent to which pre-existing immunity influences the efficacy of boosters remains unclear. In a cohort of individuals primed with the mRNA-1273 or BNT162b2 vaccines, we report that lower antibody levels before boost are associated with higher fold-increase in antibody levels after boost, suggesting that pre-existing antibody modulates the immunogenicity of mRNA vaccines. Our studies in mice show that pre-existing antibodies accelerate the clearance of vaccine antigen via Fc-dependent mechanisms, limiting the amount of antigen available to prime B cell responses after mRNA boosters. These data demonstrate a "tug of war" between pre-existing antibody responses and de novo B cell responses following mRNA vaccination, and they suggest that transient downmodulation of antibody effector function may improve the efficacy of mRNA boosters.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Min Han Lew
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Bakare Awakoaiye
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lavanya Visvabharathy
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Justin M Richner
- Department of Microbiology and Immunology, University of Illinois Chicago College of Medicine, Chicago, IL 60612, USA
| | - Igor J Koralnik
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
12
|
Einav T, Kosikova M, Radvak P, Kuo YC, Kwon HJ, Xie H. Mapping the Antibody Repertoires in Ferrets with Repeated Influenza A/H3 Infections: Is Original Antigenic Sin Really "Sinful"? Viruses 2023; 15:374. [PMID: 36851590 PMCID: PMC9959794 DOI: 10.3390/v15020374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The influenza-specific antibody repertoire is continuously reshaped by infection and vaccination. The host immune response to contemporary viruses can be redirected to preferentially boost antibodies specific for viruses encountered early in life, a phenomenon called original antigenic sin (OAS) that is suggested to be responsible for diminished vaccine effectiveness after repeated seasonal vaccination. Using a new computational tool called Neutralization Landscapes, we tracked the progression of hemagglutination inhibition antibodies within ferret antisera elicited by repeated influenza A/H3 infections and deciphered the influence of prior exposures on the de novo antibody response to evolved viruses. The results indicate that a broadly neutralizing antibody signature can nevertheless be induced by repeated exposures despite OAS induction. Our study offers a new way to visualize how immune history shapes individual antibodies within a repertoire, which may help to inform future universal influenza vaccine design.
Collapse
Affiliation(s)
- Tal Einav
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Martina Kosikova
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Peter Radvak
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Yuan-Chia Kuo
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hyung Joon Kwon
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hang Xie
- Laboratory of Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
13
|
Bartsch YC, Cizmeci D, Kang J, Zohar T, Periasamy S, Mehta N, Tolboom J, Van der Fits L, Sadoff J, Comeaux C, Callendret B, Bukreyev A, Lauffenburger DA, Bastian AR, Alter G. Antibody effector functions are associated with protection from respiratory syncytial virus. Cell 2022; 185:4873-4886.e10. [PMID: 36513064 DOI: 10.1016/j.cell.2022.11.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 08/29/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022]
Abstract
Respiratory syncytial virus (RSV) infection is a major cause of severe lower respiratory tract infection and death in young infants and the elderly. With no effective prophylactic treatment available, current vaccine candidates aim to elicit neutralizing antibodies. However, binding and neutralization have poorly predicted protection in the past, and accumulating data across epidemiologic cohorts and animal models collectively point to a role for additional antibody Fc-effector functions. To begin to define the humoral correlates of immunity against RSV, here we profiled an adenovirus 26 RSV-preF vaccine-induced humoral immune response in a group of healthy adults that were ultimately challenged with RSV. Protection from infection was linked to opsonophagocytic functions, driven by IgA and differentially glycosylated RSV-specific IgG profiles, marking a functional humoral immune signature of protection against RSV. Furthermore, Fc-modified monoclonal antibodies able to selectively recruit effector functions demonstrated significant antiviral control in a murine model of RSV.
Collapse
Affiliation(s)
- Yannic C Bartsch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jaewon Kang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Tomer Zohar
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Sivakumar Periasamy
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nickita Mehta
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeroen Tolboom
- Janssen Vaccines & Prevention BV, 2333 Leiden, the Netherlands
| | | | - Jerry Sadoff
- Janssen Vaccines & Prevention BV, 2333 Leiden, the Netherlands
| | - Christy Comeaux
- Janssen Vaccines & Prevention BV, 2333 Leiden, the Netherlands
| | | | - Alexander Bukreyev
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
14
|
Gao S, Wei M, Chu K, Li J, Zhu F. Effects of maternal antibodies in infants on the immunogenicity and safety of inactivated polio vaccine in infants. Hum Vaccin Immunother 2022; 18:2050106. [PMID: 35394898 PMCID: PMC9196670 DOI: 10.1080/21645515.2022.2050106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The presence of maternal poliovirus antibodies may interfere with the immune response to inactivated polio vaccine (IPV), and its influence on the safety of vaccination is not yet understood. A total of 1146 eligible infants were randomly assigned (1:1) to the IPV and Sabin IPV (SIPV) groups to compare and analyze the efficacy of the two vaccines in preventing poliovirus infection. We pooled the SIPV and IPV groups and reclassified them into the maternal poliovirus antibody-positive group (MAPG; ≥1: 8) and the maternal poliovirus antibody-negative group (MANG; <1: 8). We evaluated the impact of maternal poliovirus antibodies by comparing the geometric mean titer (GMT), seroconversion rate, and geometric mean increase (GMI) of types I-III poliovirus neutralizing antibodies post-vaccination, and incidence rates of adverse reactions following vaccination between the MAPG and MANG. Respective seroconversion rates in the MAPG and MANG were 94% and 100%, 79.27% and 100%, and 93.26% and 100% (all serotypes, P < .01) for types I-III poliovirus, respectively. The GMT of all types of poliovirus antibodies in the MAPG (1319.13, 219.91, 764.11, respectively) were significantly lower than those in the MANG (1584.92, 286.73, 899.59, respectively) (P < .05). The GMI in the MAPG was significantly lower than that in the MANG (P < .05). No statistically significant difference in the incidence of local and systemic adverse reactions was observed between the MAPG and MANG. Thus, the presence of maternal poliovirus antibodies does not affect the safety of IPV but can negatively impact the immune responses in infants after IPV vaccination.
Collapse
Affiliation(s)
- Shuyu Gao
- School of Public Health, Southeast University, Nanjing, PR China
| | - Mingwei Wei
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China
| | - Kai Chu
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China
| | - Jingxin Li
- Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China.,NHC Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, PR China
| | - Fengcai Zhu
- School of Public Health, Southeast University, Nanjing, PR China.,Vaccine Clinical Evaluation Department, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, PR China.,NHC Key laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, PR China
| |
Collapse
|
15
|
Wolf C, Köppert S, Becza N, Kuerten S, Kirchenbaum GA, Lehmann PV. Antibody Levels Poorly Reflect on the Frequency of Memory B Cells Generated following SARS-CoV-2, Seasonal Influenza, or EBV Infection. Cells 2022; 11:cells11223662. [PMID: 36429090 PMCID: PMC9688940 DOI: 10.3390/cells11223662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
The scope of immune monitoring is to define the existence, magnitude, and quality of immune mechanisms operational in a host. In clinical trials and praxis, the assessment of humoral immunity is commonly confined to measurements of serum antibody reactivity without accounting for the memory B cell potential. Relying on fundamentally different mechanisms, however, passive immunity conveyed by pre-existing antibodies needs to be distinguished from active B cell memory. Here, we tested whether, in healthy human individuals, the antibody titers to SARS-CoV-2, seasonal influenza, or Epstein-Barr virus antigens correlated with the frequency of recirculating memory B cells reactive with the respective antigens. Weak correlations were found. The data suggest that the assessment of humoral immunity by measurement of antibody levels does not reflect on memory B cell frequencies and thus an individual's potential to engage in an anamnestic antibody response against the same or an antigenically related virus. Direct monitoring of the antigen-reactive memory B cell compartment is both required and feasible towards that goal.
Collapse
Affiliation(s)
- Carla Wolf
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Sebastian Köppert
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Noémi Becza
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, 53115 Bonn, Germany
| | - Greg A. Kirchenbaum
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
| | - Paul V. Lehmann
- Research and Development, Cellular Technology Ltd. (CTL), Shaker Heights, OH 44122, USA
- Correspondence: ; Tel.: +1-(216)-791-5084
| |
Collapse
|
16
|
Abstract
Barrier tissues are the primary site of infection for pathogens likely to cause future pandemics. Tissue-resident lymphocytes can rapidly detect pathogens upon infection of barrier tissues and are critical in preventing viral spread. However, most vaccines fail to induce tissue-resident lymphocytes and are instead reliant on circulating antibodies to mediate protective immunity. Circulating antibody titers wane over time following vaccination leaving individuals susceptible to breakthrough infections by variant viral strains that evade antibody neutralization. Memory B cells were recently found to establish tissue residence following infection of barrier tissues. Here, we summarize emerging evidence for the importance of tissue-resident memory B cells in the establishment of protective immunity against viral and bacterial challenge. We also discuss the role of tissue-resident memory B cells in regulating the progression of non-infectious diseases. Finally, we examine new approaches to develop vaccines capable of eliciting barrier immunity.
Collapse
Affiliation(s)
- Changfeng Chen
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Brian J Laidlaw
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
17
|
Frasca D, Diaz A, Romero M, Blomberg BB. Metformin Enhances B Cell Function and Antibody Responses of Elderly Individuals With Type-2 Diabetes Mellitus. FRONTIERS IN AGING 2022; 2:715981. [PMID: 35822013 PMCID: PMC9261392 DOI: 10.3389/fragi.2021.715981] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022]
Abstract
Our previous work has shown that young and elderly patients with Type-2 Diabetes Mellitus (T2DM) treated with Metformin have optimal B cell function and serum antibodies specific for the seasonal influenza vaccine. In this paper, we have evaluated B cell function and the metabolic requirements of B cell antibody responses in elderly T2DM patients (ET2DM) taking or not Metformin, and compared to those of healthy elderly (EH) and healthy young (YH) individuals. Results show that Metformin significantly increases in vivo B cell function, measured by influenza vaccine-specific serum antibodies, in ET2DM patients to the levels observed in EH and more importantly in YH individuals. Metformin also decreases the frequencies of pro-inflammatory B cell subsets, as well as intrinsic inflammation and metabolic requirements of peripheral B cells from ET2DM. This hyper-metabolic phenotype of B cells from ET2DM is needed to support intrinsic inflammation, measured by the expression of transcripts for markers of the senescence-associated secretory phenotype (SASP), and the secretion of autoimmune antibodies. Importantly, B cell function in ET2DM patients taking Metformin is not only increased as compared to that in ET2DM patients not taking Metformin, but is comparable to B cell function measured in YH individuals. These results altogether strongly support the anti-aging effects of Metformin on humoral immunity.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
18
|
Dangi T, Sanchez S, Lew MH, Visvabharathy L, Richner J, Koralnik IJ, Penaloza-MacMaster P. Pre-existing immunity modulates responses to mRNA boosters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.06.27.497248. [PMID: 35794898 PMCID: PMC9258286 DOI: 10.1101/2022.06.27.497248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
mRNA vaccines have shown high efficacy in preventing severe COVID-19, but breakthrough infections, emerging variants and waning antibody levels have warranted the use of boosters. Although mRNA boosters have been widely implemented, the extent to which pre-existing immunity influences the efficacy of boosters remains unclear. In a cohort of individuals primed with the mRNA-1273 or BNT162b2 vaccines, we observed that lower antibody levels before boost were associated with higher fold-increase in antibody levels after boost, suggesting that pre-existing antibody modulates the boosting capacity of mRNA vaccines. Mechanistic studies in mice show that pre-existing antibodies significantly limit antigen expression and priming of B cell responses after mRNA vaccination. Furthermore, we demonstrate that the relative superiority of an updated Omicron vaccine over the original vaccine is critically dependent on the serostatus of the host. These data demonstrate that pre-existing immunity dictates responses to mRNA vaccination, elucidating specific circumstances when updated SARS-CoV-2 vaccines confer superior protection to original vaccines.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Min Han Lew
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lavanya Visvabharathy
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Justin Richner
- Department of Microbiology & Immunology, University of Illinois Chicago College of Medicine, Chicago, IL 60612
| | - Igor J. Koralnik
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
19
|
An epitope-optimized human H3N2 influenza vaccine induces broadly protective immunity in mice and ferrets. NPJ Vaccines 2022; 7:65. [PMID: 35739199 PMCID: PMC9226166 DOI: 10.1038/s41541-022-00492-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/16/2022] [Indexed: 12/03/2022] Open
Abstract
There is a crucial need for an improved H3N2 influenza virus vaccine due to low vaccine efficacy rates and increased morbidity and mortality associated with H3N2-dominated influenza seasons. Here, we utilize a computational design strategy to produce epitope-optimized, broadly cross-reactive H3 hemagglutinins in order to create a universal H3N2 influenza vaccine. The Epigraph immunogens are designed to maximize the viral population frequency of epitopes incorporated into the immunogen. We compared our Epigraph H3 vaccine to the traditional egg-based inactivated influenza vaccine from 2018-19, FluZone. Epigraph vaccination-induced stronger cross-reactive antibody responses than FluZone against 18 H3N2 viruses isolated from 1968 to 2019 in both mice and ferrets, with protective hemagglutination inhibition titers against 93-100% of the contemporary H3N2 strains compared to only 27% protection measured from FluZone. In addition, Epigraph vaccination-induced strong cross-reactive T-cell immunity which significantly contributes to protection against lethal influenza virus infection. Finally, Epigraph vaccination protected ferrets from influenza disease after challenge with two H3N2 viruses. The superior cross-reactive immunity induced by these Epigraph immunogens supports their development as a universal H3N2 influenza vaccine.
Collapse
|
20
|
Fox A, Carolan L, Leung V, Phuong HVM, Khvorov A, Auladell M, Tseng YY, Thai PQ, Barr I, Subbarao K, Mai LTQ, van Doorn HR, Sullivan SG. Opposing Effects of Prior Infection versus Prior Vaccination on Vaccine Immunogenicity against Influenza A(H3N2) Viruses. Viruses 2022; 14:470. [PMID: 35336877 PMCID: PMC8949461 DOI: 10.3390/v14030470] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/10/2021] [Accepted: 11/28/2021] [Indexed: 02/05/2023] Open
Abstract
Prior vaccination can alternately enhance or attenuate influenza vaccine immunogenicity and effectiveness. Analogously, we found that vaccine immunogenicity was enhanced by prior A(H3N2) virus infection among participants of the Ha Nam Cohort, Viet Nam, but was attenuated by prior vaccination among Australian Health Care Workers (HCWs) vaccinated in the same year. Here, we combined these studies to directly compare antibody titers against 35 A(H3N2) viruses spanning 1968-2018. Participants received licensed inactivated vaccines containing A/HongKong/4801/2014 (H3N2). The analysis was limited to participants aged 18-65 Y, and compared those exposed to A(H3N2) viruses circulating since 2009 by infection (Ha Nam) or vaccination (HCWs) to a reference group who had no recent A(H3N2) infection or vaccination (Ha Nam). Antibody responses were compared by fitting titer/titer-rise landscapes across strains, and by estimating titer ratios to the reference group of 2009-2018 viruses. Pre-vaccination, titers were lowest against 2009-2014 viruses among the reference (no recent exposure) group. Post-vaccination, titers were, on average, two-fold higher among participants with prior infection and two-fold lower among participants with 3-5 prior vaccinations compared to the reference group. Titer rise was negligible among participants with 3-5 prior vaccinations, poor among participants with 1-2 prior vaccinations, and equivalent or better among those with prior infection compared to the reference group. The enhancing effect of prior infection versus the incrementally attenuating effect of prior vaccinations suggests that these exposures may alternately promote and constrain the generation of memory that can be recalled by a new vaccine strain.
Collapse
Affiliation(s)
- Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (V.L.); (I.B.); (K.S.); (S.G.S.)
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (A.K.); (Y.-Y.T.)
| | - Louise Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (V.L.); (I.B.); (K.S.); (S.G.S.)
| | - Vivian Leung
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (V.L.); (I.B.); (K.S.); (S.G.S.)
| | - Hoang Vu Mai Phuong
- National Institute of Hygiene and Epidemiology, Ha Noi 100000, Vietnam; (H.V.M.P.); (P.Q.T.); (L.T.Q.M.)
| | - Arseniy Khvorov
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (A.K.); (Y.-Y.T.)
| | - Maria Auladell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia;
| | - Yeu-Yang Tseng
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (A.K.); (Y.-Y.T.)
| | - Pham Quang Thai
- National Institute of Hygiene and Epidemiology, Ha Noi 100000, Vietnam; (H.V.M.P.); (P.Q.T.); (L.T.Q.M.)
| | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (V.L.); (I.B.); (K.S.); (S.G.S.)
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (V.L.); (I.B.); (K.S.); (S.G.S.)
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia;
| | - Le Thi Quynh Mai
- National Institute of Hygiene and Epidemiology, Ha Noi 100000, Vietnam; (H.V.M.P.); (P.Q.T.); (L.T.Q.M.)
| | - H. Rogier van Doorn
- Oxford University Clinical Research Unit, Wellcome Africa Asia Programme, National Hospital of Tropical Diseases, Ha Noi 100000, Vietnam;
- Centre of Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LG, UK
| | - Sheena G. Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (L.C.); (V.L.); (I.B.); (K.S.); (S.G.S.)
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; (A.K.); (Y.-Y.T.)
| |
Collapse
|
21
|
Krivitskaya VZ, Kuznecova EV, Maiorova VG, Petrova ER, Sominina AA, Danilenko DM. Influenza vaccination influencing level of specific humoral immunity in healthy individuals. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2022. [DOI: 10.15789/2220-7619-ivi-1750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
To assess an effect of vaccination on the level of humoral anti-influenza herd immunity, 2955 sera were collected and analyzed by HIT in the 2019–2020 and 2020–2021 epidemiological seasons. All sera were obtained from healthy adult donors residing in various cities of the Russian Federation. Among them, 1057 volunteers were vaccinated with seasonal influenza trivalent inactivated vaccine. Characteristics of humoral anti-influenza immunity (average geometric antibody titers and the proportion of individuals seropositive for the vaccine viruses) obtained in autumn 2019 and 2020 (1–2 months after vaccination) in vaccinated individuals vs. unvaccinated subjects were found to be markedly higher evidencing about a positive vaccination-related contribution to developing herd immunity against influenza in the preepidemic periods. After the 2019–2020 influenza epidemic, in spring 2020 (6–7 months after vaccination), the levels of antibodies to all vaccine components decreased by 2.6–3.5-fold in vaccinated donors compared to the pre-epidemic period in 2019 autumn. Antibody titers became substantially lower than the protective level (titer by HIT 1/40). At the same time, no significant differences between the groups of vaccinated vs. unvaccinated individuals were observed afterwards. This indicates instability of post-vaccination anti-influenza humoral immunity. As a result, it may decrease an influenza-resistant population cohort of working age on the eve of new epidemic season. The immunogenicity of the inactivated trivalent seasonal influenza vaccine was estimated by HIT while analyzing paired sera obtained from 295 and 112 healthy individuals of various ages vaccinated in autumn 2019 and 2020, respectively. The response to the vaccine was found to be age-related. Children aged 3–14 years vs. older subjects showed a more efficient response. Insufficient immunogenicity of influenza B virus vaccine components was shown. In all age groups, average geometric titers for influenza B virus antibodies were lower (2–8-fold) than for current A(H1N1)pdm09-like strains and influenza A(H3N2) viruses 1–1.5 months post-vaccination. Analyzing vaccine immunogenicity showed a significant inverse relationship between the level of preexisting strain-specific serum antibodies before vaccination and formation of antibodies to the corresponding vaccine virus 1–1.5 months after vaccination. Seroconversion to each vaccine component was remarkably more frequent in individuals with a low preexisting level of antibodies specific to the corresponding virus.
Collapse
|
22
|
OUP accepted manuscript. Lab Med 2022; 53:602-608. [DOI: 10.1093/labmed/lmac035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
23
|
Influence of Prior Influenza Vaccination on Current Influenza Vaccine Effectiveness in Children Aged 1 to 5 Years. Vaccines (Basel) 2021; 9:vaccines9121447. [PMID: 34960193 PMCID: PMC8706378 DOI: 10.3390/vaccines9121447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/25/2021] [Accepted: 12/04/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Although annual influenza vaccination is an important strategy used to prevent influenza-related morbidity and mortality, some studies have reported the negative influence of prior vaccination on vaccine effectiveness (VE) for current seasons. Currently, the influence of prior vaccination is not conclusive, especially in children. METHODS We evaluated the association between current-season VE and prior season vaccination using a test-negative design in children aged 1-5 years presenting at nine outpatient clinics in Japan during the 2016/17 and 2017/18 influenza seasons. Children with influenza-like illness were enrolled prospectively and tested for influenza using real-time RT-PCR. Their recent vaccination history was categorized into six groups according to current vaccination doses (0/1/2) and prior vaccination status (unvaccinated = 0 doses/vaccinated = 1 dose or 2 doses): (1) 0 doses in the current season and unvaccinated in prior seasons (reference group); (2) 0 doses in the current season and vaccinated in a prior season; (3) 1 dose in the current season and unvaccinated in a prior season; (4) 1 dose in the current season and vaccinated in a prior season; (5) 2 doses in the current season and unvaccinated in a prior season, and (6) 2 doses in the current season and vaccinated in a prior season. RESULTS A total of 799 cases and 1196 controls were analyzed. The median age of the subjects was 3 years, and the proportion of males was 54%. Overall, the vaccination rates (any vaccination in the current season) in the cases and controls were 36% and 53%, respectively. The VEs of the groups were: (2) 29% (95% confidence interval: -25% to 59%); (3) 53% (6% to 76%); (4) 70% (45% to 83%); (5) 56% (32% to 72%), and (6) 61% (42% to 73%). The one- and two-dose VEs of the current season were significant regardless of prior vaccination status. The results did not differ when stratified by influenza subtype/lineage. CONCLUSION Prior vaccination did not attenuate the current-season VE in children aged 1 to 5 years, supporting the annual vaccination strategy.
Collapse
|
24
|
Hill DL, Whyte CE, Innocentin S, Lee JL, Dooley J, Wang J, James EA, Lee JC, Kwok WW, Zand MS, Liston A, Carr EJ, Linterman MA. Impaired HA-specific T follicular helper cell and antibody responses to influenza vaccination are linked to inflammation in humans. eLife 2021; 10:e70554. [PMID: 34726156 PMCID: PMC8562996 DOI: 10.7554/elife.70554] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Antibody production following vaccination can provide protective immunity to subsequent infection by pathogens such as influenza viruses. However, circumstances where antibody formation is impaired after vaccination, such as in older people, require us to better understand the cellular and molecular mechanisms that underpin successful vaccination in order to improve vaccine design for at-risk groups. Here, by studying the breadth of anti-haemagglutinin (HA) IgG, serum cytokines, and B and T cell responses by flow cytometry before and after influenza vaccination, we show that formation of circulating T follicular helper (cTfh) cells was associated with high-titre antibody responses. Using Major Histocompatability Complex (MHC) class II tetramers, we demonstrate that HA-specific cTfh cells can derive from pre-existing memory CD4+ T cells and have a diverse T cell receptor (TCR) repertoire. In older people, the differentiation of HA-specific cells into cTfh cells was impaired. This age-dependent defect in cTfh cell formation was not due to a contraction of the TCR repertoire, but rather was linked with an increased inflammatory gene signature in cTfh cells. Together, this suggests that strategies that temporarily dampen inflammation at the time of vaccination may be a viable strategy to boost optimal antibody generation upon immunisation of older people.
Collapse
Affiliation(s)
- Danika L Hill
- Department of Immunology and Pathology, Monash UniversityMelbourneAustralia
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
| | - Carly E Whyte
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
| | - Silvia Innocentin
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
| | - Jia Le Lee
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
| | - James Dooley
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
| | - Jiong Wang
- Division of Nephrology, Department of Medicine and Clinical and Translational Science Institute, University of Rochester Medical CenterRochesterUnited States
| | - Eddie A James
- Benaroya Research Institute at Virginia Mason, Translational Research Program and Tetramer Core LaboratorySeattleUnited States
| | - James C Lee
- Department of Medicine, Cambridge Biomedical Campus, University of CambridgeCambridgeUnited Kingdom
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of CambridgeCambridgeUnited Kingdom
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Diabetes ProgramSeattleUnited States
- Department of Medicine, University of WashingtonSeattleUnited States
| | - Martin S Zand
- Division of Nephrology, Department of Medicine and Clinical and Translational Science Institute, University of Rochester Medical CenterRochesterUnited States
| | - Adrian Liston
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
| | - Edward J Carr
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
- Department of Medicine, Cambridge Biomedical Campus, University of CambridgeCambridgeUnited Kingdom
| | - Michelle A Linterman
- Immunology Program, The Babraham Institute, Babraham Research CampusCambridgeUnited Kingdom
| |
Collapse
|
25
|
Walti CS, Loes AN, Shuey K, Krantz EM, Boonyaratanakornkit J, Keane-Candib J, Loeffelholz T, Wolf CR, Taylor JJ, Gardner RA, Green DJ, Cowan AJ, Maloney DG, Turtle CJ, Pergam SA, Chu HY, Bloom JD, Hill JA. Humoral immunogenicity of the seasonal influenza vaccine before and after CAR-T-cell therapy: a prospective observational study. J Immunother Cancer 2021; 9:e003428. [PMID: 34702753 PMCID: PMC8549667 DOI: 10.1136/jitc-2021-003428] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 11/23/2022] Open
Abstract
Recipients of chimeric antigen receptor-modified T (CAR-T) cell therapies for B cell malignancies have profound and prolonged immunodeficiencies and are at risk for serious infections, including respiratory virus infections. Vaccination may be important for infection prevention, but there are limited data on vaccine immunogenicity in this population. We conducted a prospective observational study of the humoral immunogenicity of commercially available 2019-2020 inactivated influenza vaccines in adults immediately prior to or while in durable remission after CD19-, CD20-, or B cell maturation antigen-targeted CAR-T-cell therapy, as well as controls. We tested for antibodies to all four vaccine strains using neutralization and hemagglutination inhibition (HAI) assays. Antibody responses were defined as at least fourfold titer increases from baseline. Seroprotection was defined as a HAI titer ≥40. Enrolled CAR-T-cell recipients were vaccinated 14-29 days prior to (n=5) or 13-57 months following therapy (n=13), and the majority had hypogammaglobulinemia and cellular immunodeficiencies prevaccination. Eight non-immunocompromised adults served as controls. Antibody responses to ≥1 vaccine strain occurred in 2 (40%) individuals before CAR-T-cell therapy and in 4 (31%) individuals vaccinated after CAR-T-cell therapy. An additional 1 (20%) and 6 (46%) individuals had at least twofold increases, respectively. One individual vaccinated prior to CAR-T-cell therapy maintained a response for >3 months following therapy. Across all tested vaccine strains, seroprotection was less frequent in CAR-T-cell recipients than in controls. There was evidence of immunogenicity even among individuals with low immunoglobulin, CD19+ B cell, and CD4+ T-cell counts. These data support consideration for vaccination before and after CAR-T-cell therapy for influenza and other relevant pathogens such as SARS-CoV-2, irrespective of hypogammaglobulinemia or B cell aplasia. However, relatively impaired humoral vaccine immunogenicity indicates the need for additional infection-prevention strategies. Larger studies are needed to refine our understanding of potential correlates of vaccine immunogenicity, and durability of immune responses, in CAR-T-cell therapy recipients.
Collapse
Affiliation(s)
- Carla S Walti
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andrea N Loes
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Howard Hughes Medical Institute, Seattle, Washington, USA
| | - Kiel Shuey
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Elizabeth M Krantz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jacob Keane-Candib
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Tillie Loeffelholz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Caitlin R Wolf
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Rebecca A Gardner
- Division of Hematology-Oncology, Seattle Children's Hospital, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Damian J Green
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andrew J Cowan
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - David G Maloney
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Cameron J Turtle
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Steven A Pergam
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Helen Y Chu
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Jesse D Bloom
- Howard Hughes Medical Institute, Seattle, Washington, USA
- Basic Sciences Division, Computational Biology Program, and Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joshua A Hill
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Vaccine and Infectious Disease Division, and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
26
|
Abstract
Innate and adaptive immune responses decline with age, leading to greater susceptibility to infectious diseases and reduced responses to vaccines. Diseases are more severe in old than in young individuals and have a greater impact on health outcomes such as morbidity, disability, and mortality. Aging is characterized by increased low-grade chronic inflammation, so-called inflammaging, that represents a link between changes in immune cells and a number of diseases and syndromes typical of old age. In this review we summarize current knowledge on age-associated changes in immune cells with special emphasis on B cells, which are more inflammatory and less responsive to infections and vaccines in the elderly. We highlight recent findings on factors and pathways contributing to inflammaging and how these lead to dysfunctional immune responses. We summarize recent published studies showing that adipose tissue, which increases in size with aging, contributes to inflammaging and dysregulated B cell function.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA; .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA.,Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA;
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA;
| | - Denisse Garcia
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA;
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA; .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
27
|
Wen S, Wu Z, Zhong S, Li M, Shu Y. Factors influencing the immunogenicity of influenza vaccines. Hum Vaccin Immunother 2021; 17:2706-2718. [PMID: 33705263 DOI: 10.1080/21645515.2021.1875761] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Annual vaccination is the best prevention of influenza. However, the immunogenicity of influenza vaccines varies among different populations. It is important to fully identify the factors that may affect the immunogenicity of the vaccines to provide best protection for vaccine recipients. This paper reviews the factors that may influence the immunogenicity of influenza vaccines from the aspects of vaccine factors, adjuvants, individual factors, repeated vaccination, and genetic factors. The confirmed or hypothesized molecular mechanisms of these factors have also been briefly summarized.
Collapse
Affiliation(s)
- Simin Wen
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Zhengyu Wu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Shuyi Zhong
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Mao Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China.,National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Prevention and Control, Beijing, China
| |
Collapse
|
28
|
Rouphael NG, Lai L, Tandon S, McCullough MP, Kong Y, Kabbani S, Natrajan MS, Xu Y, Zhu Y, Wang D, O'Shea J, Sherman A, Yu T, Henry S, McAllister D, Stadlbauer D, Khurana S, Golding H, Krammer F, Mulligan MJ, Prausnitz MR. Immunologic mechanisms of seasonal influenza vaccination administered by microneedle patch from a randomized phase I trial. NPJ Vaccines 2021; 6:89. [PMID: 34262052 PMCID: PMC8280206 DOI: 10.1038/s41541-021-00353-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
In a phase 1 randomized, single-center clinical trial, inactivated influenza virus vaccine delivered through dissolvable microneedle patches (MNPs) was found to be safe and immunogenic. Here, we compare the humoral and cellular immunologic responses in a subset of participants receiving influenza vaccination by MNP to the intramuscular (IM) route of administration. We collected serum, plasma, and peripheral blood mononuclear cells in 22 participants up to 180 days post-vaccination. Hemagglutination inhibition (HAI) titers and antibody avidity were similar after MNP and IM vaccination, even though MNP vaccination used a lower antigen dose. MNPs generated higher neuraminidase inhibition (NAI) titers for all three influenza virus vaccine strains tested and triggered a larger percentage of circulating T follicular helper cells (CD4 + CXCR5 + CXCR3 + ICOS + PD-1+) compared to the IM route. Our study indicates that inactivated influenza virus vaccination by MNP produces humoral and cellular immune response that are similar or greater than IM vaccination.
Collapse
Affiliation(s)
- Nadine G Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia.
| | - Lilin Lai
- Emory Vaccine Center, Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia
| | - Sonia Tandon
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia.,Laney Graduate School, Emory University, Atlanta, Georgia
| | - Michele Paine McCullough
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Yunchuan Kong
- Laney Graduate School, Emory University, Atlanta, Georgia
| | - Sarah Kabbani
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Muktha S Natrajan
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Yerun Zhu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Dongli Wang
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Jesse O'Shea
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Amy Sherman
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Tianwei Yu
- Laney Graduate School, Emory University, Atlanta, Georgia
| | | | | | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Surender Khurana
- Division of Viral Products Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Hana Golding
- Division of Viral Products Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark J Mulligan
- New York University Langone Medical Center, Alexandria Center for Life Sciences, New York, NY, USA
| | - Mark R Prausnitz
- Micron Biomedical, Inc., Atlanta, Georgia.,School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
29
|
Guthmiller JJ, Utset HA, Wilson PC. B Cell Responses against Influenza Viruses: Short-Lived Humoral Immunity against a Life-Long Threat. Viruses 2021; 13:965. [PMID: 34067435 PMCID: PMC8224597 DOI: 10.3390/v13060965] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/25/2022] Open
Abstract
Antibodies are critical for providing protection against influenza virus infections. However, protective humoral immunity against influenza viruses is limited by the antigenic drift and shift of the major surface glycoproteins, hemagglutinin and neuraminidase. Importantly, people are exposed to influenza viruses throughout their life and tend to reuse memory B cells from prior exposure to generate antibodies against new variants. Despite this, people tend to recall memory B cells against constantly evolving variable epitopes or non-protective antigens, as opposed to recalling them against broadly neutralizing epitopes of hemagglutinin. In this review, we discuss the factors that impact the generation and recall of memory B cells against distinct viral antigens, as well as the immunological limitations preventing broadly neutralizing antibody responses. Lastly, we discuss how next-generation vaccine platforms can potentially overcome these obstacles to generate robust and long-lived protection against influenza A viruses.
Collapse
Affiliation(s)
- Jenna J. Guthmiller
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
| | - Henry A. Utset
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
| | - Patrick C. Wilson
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (H.A.U.); (P.C.W.)
- Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
30
|
Walti CS, Loes AN, Shuey K, Krantz EM, Boonyaratanakornkit J, Keane-Candib J, Loeffelholz T, Wolf CR, Taylor JJ, Gardner RA, Green DJ, Cowan AJ, Maloney DG, Turtle CJ, Pergam SA, Chu HY, Bloom JD, Hill JA. Humoral immunogenicity of the seasonal influenza vaccine before and after CAR-T-cell therapy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34013294 PMCID: PMC8132269 DOI: 10.1101/2021.05.10.21256634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recipients of chimeric antigen receptor-modified T (CAR-T) cell therapies for B-cell malignancies are immunocompromised and at risk for serious infections. Vaccine immunogenicity is unknown in this population. We conducted a prospective observational study of the humoral immunogenicity of 2019-2020 inactivated influenza vaccines (IIV) in children and adults immediately prior to (n=7) or 13-57 months after (n=15) CD19-, CD20-, or BCMA-targeted CAR-T-cell therapy, as well as controls (n=8). Individuals post-CAR-T-cell therapy were in remission. We tested for antibodies to 4 vaccine strains at baseline and ≥1 time point after IIV using neutralization and hemagglutination inhibition assays. An antibody response was defined as a ≥4-fold titer increase from baseline at the first post-vaccine time point. Baseline A(H1N1) titers in the CAR-T cohorts were significantly lower compared to controls. Antibody responses to ≥1 vaccine strain occurred in 2 (29%) individuals before CAR-T-cell therapy; one individual maintained a response for >3 months post-CAR-T-cell therapy. Antibody responses to ≥1 vaccine strain occurred in 6 (40%) individuals vaccinated after CAR-T-cell therapy. An additional 2 (29%) and 6 (40%) individuals had ≥2-fold increases (at any time) in the pre- and post-CAR-T cohorts, respectively. There were no identified clinical or immunologic predictors of antibody responses. Neither severe hypogammaglobulinemia nor B-cell aplasia precluded antibody responses. These data support consideration for vaccination before and after CAR-T-cell therapy for influenza and other relevant pathogens such as SARS-CoV-2, irrespective of hypogammaglobulinemia or B-cell aplasia. Larger studies are needed to determine correlates of vaccine immunogenicity and durability in CAR-T-cell therapy recipients. Key Points Influenza vaccination was immunogenic pre- and post-CAR-T-cell therapy, despite hypogammaglobulinemia and B-cell aplasia.Vaccination with inactivated vaccines can be considered before CAR-T-cell therapy and in individuals with remission after therapy.
Collapse
|
31
|
Abstract
We review the phenomenon of "original antigenic sin" (OAS) in antibody responses to influenza A virus (IAV) infection or vaccination. OAS refers to the preferential induction of antibodies with higher affinity to priming versus boosting immunogens. We emphasize its mechanistic basis and origins in the basic immunobiology of B-cell responses to myriad immunogens. We tabulate 23 studies in animals and humans to show that the magnitude of OAS depends on many variables. We discuss a number of misconceptions about OAS, examine the extent to which OAS is sinful, and argue that OAS is evolutionary selected and not a deleterious by-product of selection for other features of the immune response. We end by raising questions regarding the mechanistic basis of OAS whose answers could contribute to improving influenza virus vaccines on the road to the holy grail of a "universal" influenza vaccine.
Collapse
Affiliation(s)
- Jonathan W Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jefferson J S Santos
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
32
|
Dhar N, Kwatra G, Nunes MC, Cutland C, Izu A, Nachbagauer R, Krammer F, Madhi SA. Hemagglutinin Stalk Antibody Responses Following Trivalent Inactivated Influenza Vaccine Immunization of Pregnant Women and Association With Protection From Influenza Virus Illness. Clin Infect Dis 2021; 71:1072-1079. [PMID: 31565750 PMCID: PMC7428398 DOI: 10.1093/cid/ciz927] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 09/17/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The conserved, immuno-subdominant influenza virus hemagglutinin (HA) stalk region is a potential universal group-specific influenza virus vaccine epitope. We analyzed antibody responses to H1 hemagglutinin stalk domain (H1/stalk) following trivalent influenza inactivated vaccine (IIV3) immunization in pregnant women, and association with protection against influenza virus illness. METHODS One hundred forty-five human immunodeficiency virus (HIV)-uninfected pregnant women (68 IIV3 and 77 placebo recipients) and 140 pregnant women with HIV infection (72 IIV3 and 68 placebo recipients) were independently randomized in placebo-controlled efficacy trials of IIV3. Plasma samples were tested for H1/stalk immunoglobulin G (IgG) and hemagglutination inhibition (HAI) antibodies prevaccination and 1 month postvaccination. Women had weekly surveillance for influenza illness, confirmed by polymerase chain reaction. RESULTS Increases in H1/stalk IgG (and HAI) antibody levels were elicited post-IIV3, with responses being higher in HIV-uninfected women than in women living with HIV. Among HIV-uninfected vaccinees, there was no correlation (postvaccination) between H1/stalk and HAI antibody responses, whereas a strong correlation was observed in vaccinees with HIV. The H1/stalk IgG concentration was lower among women developing A/H1N1 illness (85.3 arbitrary units [AU]/mL) than those without A/H1N1 illness (219.6 AU/mL; P = .001). H1/stalk IgG concentration ≥215 AU/mL was associated with 90% lower odds (odds ratio, 0.09; P = .005) of A/H1N1 illness. Also, H1/stalk IgG was significantly lower among women with influenza B illness (93.9 AU/mL) than among their counterparts (215.5 AU/mL) (P = .04); however, no association was observed after adjusting for HAI titers. CONCLUSIONS H1/stalk IgG concentration was associated with lower odds for A/H1N1 influenza virus illness, indicating its potential as an epitope for a universal vaccine against group 1 influenza virus.
Collapse
Affiliation(s)
- Nisha Dhar
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology, National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology, National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marta C Nunes
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology, National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Clare Cutland
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology, National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Alane Izu
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology, National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Shabir A Madhi
- Medical Research Council, Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology, National Research Foundation, Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
33
|
Balanced Cellular and Humoral Immune Responses Targeting Multiple Antigens in Adults Receiving a Quadrivalent Inactivated Influenza Vaccine. Vaccines (Basel) 2021; 9:vaccines9050426. [PMID: 33922875 PMCID: PMC8146362 DOI: 10.3390/vaccines9050426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 11/25/2022] Open
Abstract
The role of T cell immunity has been acknowledged in recent vaccine development and evaluation. We tested the humoral and cellular immune responses to Flucelvax®, a quadrivalent inactivated seasonal influenza vaccine containing two influenza A (H1N1 Singapore/GP1908/2015 IVR-180 and H3N2 North Carolina/04/2016) and two influenza B (Iowa/06/2017 and Singapore/INFTT-16-0610/2016) virus strains, using peripheral blood mononuclear cells stimulated by pools of peptides overlapping all the individual influenza viral protein components. Baseline reactivity was detected against all four strains both at the level of CD4 and CD8 responses and targeting different proteins. CD4 T cell reactivity was mostly directed to HA/NA proteins in influenza B strains, and NP/M1/M2/NS1/NEP proteins in the case of the Influenza A strains. CD8 responses to both influenza A and B viruses preferentially targeted the more conserved core viral proteins. Following vaccination, both CD4 and CD8 responses against the various influenza antigens were increased in day 15 to day 91 post vaccination period, and maintained a Th1 polarized profile. Importantly, no vaccine interference was detected, with the increased responses balanced across all four included viral strains for both CD4 and CD8 T cells, and targeting HA and multiple additional viral antigens.
Collapse
|
34
|
de Armas LR, George V, Filali-Mouhim A, Steel C, Parmigiani A, Cunningham CK, Weinberg A, Trautmann L, Sekaly RP, Cameron MJ, Pahwa S. Transcriptional and Immunologic Correlates of Response to Pandemic Influenza Vaccine in Aviremic, HIV-Infected Children. Front Immunol 2021; 12:639358. [PMID: 33868267 PMCID: PMC8044856 DOI: 10.3389/fimmu.2021.639358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/09/2021] [Indexed: 11/17/2022] Open
Abstract
People living with HIV (PWH) often exhibit poor responses to influenza vaccination despite effective combination anti-retroviral (ART) mediated viral suppression. There exists a paucity of data in identifying immune correlates of influenza vaccine response in context of HIV infection that would be useful in improving its efficacy in PWH, especially in younger individuals. Transcriptomic data were obtained by microarray from whole blood isolated from aviremic pediatric and adolescent HIV-infected individuals (4-25 yrs) given two doses of Novartis/H1N1 09 vaccine during the pandemic H1N1 influenza outbreak. Supervised clustering and gene set enrichment identified contrasts between individuals exhibiting high and low antibody responses to vaccination. High responders exhibited hemagglutination inhibition antibody titers >1:40 post-first dose and 4-fold increase over baseline. Baseline molecular profiles indicated increased gene expression in metabolic stress pathways in low responders compared to high responders. Inflammation-related and interferon-inducible gene expression pathways were higher in low responders 3 wks post-vaccination. The broad age range and developmental stage of participants in this study prompted additional analysis by age group (e.g. <13yrs and ≥13yrs). This analysis revealed differential enrichment of gene pathways before and after vaccination in the two age groups. Notably, CXCR5, a homing marker expressed on T follicular helper (Tfh) cells, was enriched in high responders (>13yrs) following vaccination which was accompanied by peripheral Tfh expansion. Our results comprise a valuable resource of immune correlates of vaccine response to pandemic influenza in HIV infected children that may be used to identify favorable targets for improved vaccine design in different age groups.
Collapse
Affiliation(s)
- Lesley R de Armas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Varghese George
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Courtney Steel
- Collaborative Genomics Center, Vaccine and Gene Therapy Institute, Port St. Lucie, FL, United States
| | - Anita Parmigiani
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Coleen K Cunningham
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Adriana Weinberg
- Departments of Medicine, Pathology, and Pediatric Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lydie Trautmann
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Rafick-Pierre Sekaly
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
35
|
Liao K, Chen K, Brett S, Gehman A, Schwartz AM, Gunn GR, DeWall SL. Characterization of the robust humoral immune response to GSK2618960, a humanized anti-IL-7 receptor monoclonal antibody, observed in healthy subjects in a Phase 1 study. PLoS One 2021; 16:e0249049. [PMID: 33755713 PMCID: PMC7987154 DOI: 10.1371/journal.pone.0249049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/08/2021] [Indexed: 12/25/2022] Open
Abstract
Interleukin-7 (IL-7) signaling modulates T cell activity and is implicated in numerous autoimmune diseases. An anti-IL-7 receptor monoclonal antibody (GSK2618960) biotherapeutic was evaluated in healthy subjects for safety, pharmacokinetics (PK), pharmacodynamics (PD) and immunogenicity in a single-dose escalation phase I study. We found that antibodies against GSK2618960 (i.e., anti-drug antibodies or ADA) developed in 83% and 100% of GSK2618960-treated subjects in the 0.6 and 2.0 mg/kg dose cohorts, respectively. Of the ADA positive subjects, 64% (7 of 11) had detectable neutralizing activity. Further investigation revealed the presence of GSK2618960-specific memory B cells, indicating the development of immunological memory for the ADAs. Ex vivo stimulation of peripheral blood mononuclear cell (PBMC) samples demonstrated a relatively strong CD4+ T cell proliferation response to GSK2618960 as compared to the control anti-RSV antibody (which is known to have only low immunogenic potential), confirming the high immunogenic potential of GSK2618960. Furthermore, GSK2618960 was found to bind in vitro monocyte-derived dendritic cells (DCs). GSK2618960 treatment of PBMCs increased the proportion of DC cells showing an increase in expression of CD83, CD86 and CD209, which indicated enhanced DC differentiation and activation relative to the isotype control anti-β amyloid antibody. Collectively, the evidence supports that the high incidence of observed clinical immunogenicity was likely related to the receptor-mediated activity by GSK2618960.
Collapse
Affiliation(s)
- Karen Liao
- Immunogenicity Group, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Keguan Chen
- Immunogenicity Group, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Sara Brett
- Oncology Cell Therapy, Oncology R&D, Stevenage, United Kingdom
| | - Andrew Gehman
- Research Statistics, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Ann M Schwartz
- Immunogenicity Group, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - George R Gunn
- Immunogenicity Group, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Stephen L DeWall
- Immunogenicity Group, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| |
Collapse
|
36
|
Forgacs D, Abreu RB, Sautto GA, Kirchenbaum GA, Drabek E, Williamson KS, Kim D, Emerling DE, Ross TM. Convergent antibody evolution and clonotype expansion following influenza virus vaccination. PLoS One 2021; 16:e0247253. [PMID: 33617543 PMCID: PMC7899375 DOI: 10.1371/journal.pone.0247253] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in high-throughput single cell sequencing have opened up new avenues into the investigation of B cell receptor (BCR) repertoires. In this study, PBMCs were collected from 17 human participants vaccinated with the split-inactivated influenza virus vaccine during the 2016-2017 influenza season. A combination of Immune Repertoire Capture (IRCTM) technology and IgG sequencing was performed on ~7,800 plasmablast (PB) cells and preferential IgG heavy-light chain pairings were investigated. In some participants, a single expanded clonotype accounted for ~22% of their PB BCR repertoire. Approximately 60% (10/17) of participants experienced convergent evolution, possessing public PBs that were elicited independently in multiple participants. Binding profiles of one private and three public PBs confirmed they were all subtype-specific, cross-reactive hemagglutinin (HA) head-directed antibodies. Collectively, this high-resolution antibody repertoire analysis demonstrated the impact evolution can have on BCRs in response to influenza virus vaccination, which can guide future universal influenza prophylactic approaches.
Collapse
Affiliation(s)
- David Forgacs
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Rodrigo B. Abreu
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Giuseppe A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Greg A. Kirchenbaum
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Elliott Drabek
- Atreca, Inc., South San Francisco, CA, United States of America
| | | | - Dongkyoon Kim
- Atreca, Inc., South San Francisco, CA, United States of America
| | | | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States of America
- * E-mail:
| |
Collapse
|
37
|
Roy S, Williams CM, Wijesundara DK, Furuya Y. Impact of Pre-Existing Immunity to Influenza on Live-Attenuated Influenza Vaccine (LAIV) Immunogenicity. Vaccines (Basel) 2020; 8:E683. [PMID: 33207559 PMCID: PMC7711626 DOI: 10.3390/vaccines8040683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022] Open
Abstract
During the previous influenza seasons, between 2010 and 2016, the live attenuated influenza vaccine (LAIV) provided variable efficacy against influenza in the U.S., causing the recommendation against the use of the LAIV. In striking contrast, pre-clinical studies have repeatedly demonstrated superior efficacy of LAIV against mismatched influenza viruses, compared to inactivated influenza vaccines (IIV). This disparity in reported vaccine efficacies between pre-clinical and clinical studies may in part be explained by limitations of the animal models of influenza. In particular, the absence of pre-existing immunity in animal models has recently emerged as a potential explanation for the discrepancies between preclinical findings and human studies. This commentary focuses on the potential impact of pre-existing immunity on LAIV induced immunogenicity with an emphasis on cross-protective immunity.
Collapse
Affiliation(s)
- Sreeja Roy
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| | - Clare M. Williams
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| | - Danushka K. Wijesundara
- The School of Chemistry and Molecular Biosciences, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland 4072, Australia;
| | - Yoichi Furuya
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| |
Collapse
|
38
|
Recombinant HA-based vaccine outperforms split and subunit vaccines in elicitation of influenza-specific CD4 T cells and CD4 T cell-dependent antibody responses in humans. NPJ Vaccines 2020; 5:77. [PMID: 32884842 PMCID: PMC7450042 DOI: 10.1038/s41541-020-00227-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Although traditional egg-based inactivated influenza vaccines can protect against infection, there have been significant efforts to develop improved formats to overcome disadvantages of this platform. Here, we have assessed human CD4 T cell responses to a traditional egg-based influenza vaccine with recently available cell-derived vaccines and recombinant baculovirus-derived vaccines. Adults were administered either egg-derived Fluzone®, mammalian cell-derived Flucelvax® or recombinant HA (Flublok®). CD4 T cell responses to each HA protein were assessed by cytokine EliSpot and intracellular staining assays. The specificity and magnitude of antibody responses were quantified by ELISA and HAI assays. By all criteria, Flublok vaccine exhibited superior performance in eliciting both CD4 T cell responses and HA-specific antibody responses, whether measured by mean response magnitude or percent of responders. Although the mechanism(s) underlying this advantage is not yet clear, it is likely that both qualitative and quantitative features of the vaccines impact the response.
Collapse
|
39
|
Wang W, Chen Q, Ford-Siltz LA, Katzelnick LC, Parra GI, Song HS, Vassell R, Weiss CD. Neutralizing Antibody Responses to Homologous and Heterologous H1 and H3 Influenza A Strains After Vaccination With Inactivated Trivalent Influenza Vaccine Vary With Age and Prior-year Vaccination. Clin Infect Dis 2020; 68:2067-2078. [PMID: 30256912 DOI: 10.1093/cid/ciy818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/21/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Prior influenza immunity influences the homologous neutralizing antibody responses elicited by inactivated influenza vaccines (IIV), but neutralizing antibody responses to heterologous strains have not been extensively characterized. METHODS We analyzed neutralizing antibody titers in individuals aged 1-88 who received the 2009-2010 season IIV before infection by or vaccination against the 2009 pandemic H1N1 virus. Neutralization titers to homologous and heterologous past, recent, and advanced H1 and H3 strains, as well as H2, H5, and H7 strains, were measured using influenza hemagglutinin pseudoviruses. We performed exploratory analyses based on age, prior-year IIV, and prevaccination titer, without controlling for Type I errors. RESULTS IIV elicited neutralizing antibodies to past and advanced H1 and H3 strains, as well as to an H2 strain in individuals who were likely infected early in life. The neutralization of avian subtype viruses was rare, and there was no imprinting of neutralization responses to novel avian subtype viruses based on the influenza group. Compared to adults, children had higher seroresponse rates to homologous and heterologous strains, and their sera generated larger antigenic distances among strains. Seroresponse rates to homologous and heterologous strains were lower in subjects vaccinated with prior-year IIV, though postimmunization titers were generally high. CONCLUSIONS IIV elicited neutralizing antibodies to heterologous H1 and H3 strains in all ages groups, but titers and seroresponse rates were usually higher in children. Prior-year vaccination with the same strains tended to blunt IIV neutralization responses to all strains in young and old age groups, yet postimmunization titers were high.
Collapse
Affiliation(s)
- Wei Wang
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Qiong Chen
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Lauren A Ford-Siltz
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Leah C Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley
| | - Gabriel I Parra
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Hyo Sook Song
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Russell Vassell
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Carol D Weiss
- Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
40
|
Frasca D, Blomberg BB, Garcia D, Keilich SR, Haynes L. Age-related factors that affect B cell responses to vaccination in mice and humans. Immunol Rev 2020; 296:142-154. [PMID: 32484934 DOI: 10.1111/imr.12864] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/16/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022]
Abstract
Aging significantly changes the ability to respond to vaccinations and infections. In this review, we summarize published results on age-related changes in response to infection with the influenza virus and on the factors known to increase influenza risk infection leading to organ failure and death. We also summarize how aging affects the response to the influenza vaccine with a special focus on B cells, which have been shown to be less responsive in the elderly. We show the cellular and molecular mechanisms contributing to the dysfunctional immune response of the elderly to the vaccine against influenza. These include a defective interaction of helper T cells (CD4+) with B cells in germinal centers, changes in the microenvironment, and the generation of immune cells with a senescence-associated phenotype. Finally, we discuss the effects of aging on metabolic pathways and we show how metabolic complications associated with aging lead to immune dysfunction.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Denisse Garcia
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Spencer R Keilich
- UConn Center on Aging, Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Laura Haynes
- UConn Center on Aging, Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
41
|
The Effects of Pre-Existing Antibodies on Live-Attenuated Viral Vaccines. Viruses 2020; 12:v12050520. [PMID: 32397218 PMCID: PMC7290594 DOI: 10.3390/v12050520] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Live-attenuated vaccines (LAVs) have achieved remarkable successes in controlling virus spread, as well as for other applications such as cancer immunotherapy. However, with rapid increases in international travel, globalization, geographic spread of viral vectors, and widespread use of vaccines, there is an increasing need to consider how pre-exposure to viruses which share similar antigenic regions can impact vaccine efficacy. Pre-existing antibodies, derived from either from maternal–fetal transmission, or by previous infection or vaccination, have been demonstrated to interfere with vaccine immunogenicity of measles, adenovirus, and influenza LAVs. Immune interference of LAVs can be caused by the formation of virus–antibody complexes that neutralize virus infection in antigen-presenting cells, or by the cross-linking of the B-cell receptor with the inhibitory receptor, FcγRIIB. On the other hand, pre-existing antibodies can augment flaviviral LAV efficacy such as that of dengue and yellow fever virus, especially when pre-existing antibodies are present at sub-neutralizing levels. The increased vaccine immunogenicity can be facilitated by antibody-dependent enhancement of virus infection, enhancing virus uptake in antigen-presenting cells, and robust induction of innate immune responses that promote vaccine immunogenicity. This review examines the literature on this topic and examines the circumstances where pre-existing antibodies can inhibit or enhance LAV efficacy. A better knowledge of the underlying mechanisms involved could allow us to better manage immunization in seropositive individuals and even identify possibilities that could allow us to exploit pre-existing antibodies to boost vaccine-induced responses for improved vaccine efficacy.
Collapse
|
42
|
Alcorn JF, Avula R, Chakka AB, Schwarzmann WE, Nowalk MP, Lin CJ, Ortiz MA, Horne WT, Chandran UR, Nagg JP, Zimmerman RK, Cole KS, Moehling KK, Martin JM. Differential gene expression in peripheral blood mononuclear cells from children immunized with inactivated influenza vaccine. Hum Vaccin Immunother 2020; 16:1782-1790. [PMID: 32298194 DOI: 10.1080/21645515.2020.1711677] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human immune response to inactivated influenza vaccine is dynamic and impacted by age and preexisting immunity. Our goal was to identify postvaccination transcriptomic changes in peripheral blood mononuclear cells from children. Blood samples were obtained before and at 3 or 7 days postvaccination with 2016-2017 quadrivalent inactivated influenza vaccine and RNA sequencing was performed. There were 1,466 differentially expressed genes (DEGs) for the Day 0-Day 3 group and 513 DEGs for the Day 0-Day 7 group. Thirty-three genes were common between the two groups. The majority of the transcriptomic changes at Day 3 represented innate inflammation and apoptosis pathways. Day 7 DEGs were characterized by activation of cellular processes, including the regulation of cytoskeleton, junctions, and metabolism, and increased expression of immunoglobulin genes. DEGs at Day 3 were compared between older and younger children revealing increased inflammatory gene expression in the older group. Vaccine history in the year prior to the study was characterized by robust DEGs at Day 3 with decreased phagosome and dendritic cell maturation in those who had been vaccinated in the previous year. PBMC responses to inactivated influenza vaccination in children differed significantly by the timing of sampling, patient age, and vaccine history. These data provide insight into the expected molecular pathways to be temporally altered by influenza vaccination in children.
Collapse
Affiliation(s)
- John F Alcorn
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Raghunandan Avula
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Anish B Chakka
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | - William E Schwarzmann
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | | | | | - Marianna A Ortiz
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA.,Center for Vaccine Research, University of Pittsburgh , Pittsburgh, PA, USA
| | - William T Horne
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Uma R Chandran
- Department of Biomedical Informatics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Jennifer P Nagg
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| | - Richard K Zimmerman
- Department of Family Medicine, University of Pittsburgh , Pittsburgh, PA, USA
| | - Kelly S Cole
- Center for Vaccine Research, University of Pittsburgh , Pittsburgh, PA, USA
| | - Krissy K Moehling
- Department of Family Medicine, University of Pittsburgh , Pittsburgh, PA, USA
| | - Judith M Martin
- Department of Pediatrics, University of Pittsburgh , Pittsburgh, PA, USA
| |
Collapse
|
43
|
Baseline Levels of Influenza-Specific B Cells and T Cell Responses Modulate Human Immune Responses to Swine Variant Influenza A/H3N2 Vaccine. Vaccines (Basel) 2020; 8:vaccines8010126. [PMID: 32183105 PMCID: PMC7157591 DOI: 10.3390/vaccines8010126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 11/16/2022] Open
Abstract
The cellular immune responses elicited by an investigational vaccine against an emergent variant of influenza (H3N2v) are not fully understood. Twenty-five subjects, enrolled in an investigational influenza A/H3N2v vaccine study, who received two doses of vaccine 21 days apart, were included in a sub-study of cellular immune responses. H3N2v-specific plasmablasts were determined by ELISpot 8 days after each vaccine dose and H3N2v specific CD4+ T cells were quantified by intracellular cytokine and CD154 (CD40 ligand) staining before vaccination, 8 and 21 days after each vaccine dose. Results: 95% (19/20) and 96% (24/25) subjects had pre-existing H3N2v specific memory B, and T cell responses, respectively. Plasmablast responses at Day 8 after the first vaccine administration were detected against contemporary H3N2 strains and correlated with hemagglutination inhibition HAI (IgG: p = 0.018; IgA: p < 0.001) and Neut (IgG: p = 0.038; IgA: p = 0.021) titers and with memory B cell frequency at baseline (IgA: r = 0.76, p < 0.001; IgG: r = 0.74, p = 0.0001). The CD4+ T cells at Days 8 and 21 expanded after prime vaccination and this expansion correlated strongly with early post-vaccination HAI and Neut titers (p ≤ 0.002). In an adult population, the rapid serological response observed after initial H3N2v vaccination correlates with post-vaccination plasmablasts and CD4+ T cell responses.
Collapse
|
44
|
Matrajt L, Halloran ME, Antia R. Successes and Failures of the Live-attenuated Influenza Vaccine: Can We Do Better? Clin Infect Dis 2020; 70:1029-1037. [PMID: 31056675 PMCID: PMC7319054 DOI: 10.1093/cid/ciz358] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/30/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The effectiveness of the live-attenuated influenza vaccine (LAIV) can vary widely, ranging from 0% to 50%. The reasons for these discrepancies remain largely unclear. METHODS We use mathematical models to explore how the efficacy of LAIV is affected by the degree of mismatch with the currently circulating influenza strain and interference with pre-existing immunity. The models incorporate 3 key antigenic distances: the distances between the vaccine strain, pre-existing immunity, and the challenge strain. RESULTS Our models show that an LAIV that is matched with the currently circulating strain is likely to have only modest efficacy. Our results suggest that the efficacy of the vaccine would be increased (optimized) if, rather than being matched to the circulating strain, it is antigenically slightly further from pre-existing immunity than the circulating strain. The models also suggest 2 regimes in which LAIV that is matched to circulating strains may be protective: in children before they have built immunity to circulating strains and in response to novel strains (such as antigenic shifts) which are at substantial antigenic distance from previously circulating strains. We provide an explanation for the variation in vaccine effectiveness between studies and countries of vaccine effectiveness observed during the 2014-2015 influenza season. CONCLUSIONS LAIV is offered to children across the world; however, its effectiveness significantly varies between studies. Here, we propose a mechanistic explanation to understand these differences. We further propose a way to select the LAIV strain that would have a higher chance of being protective.
Collapse
Affiliation(s)
- Laura Matrajt
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
| | - M Elizabeth Halloran
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center
- Department of Biostatistics, University of Washington, Seattle
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, Georgia
| |
Collapse
|
45
|
Zelner J, Petrie JG, Trangucci R, Martin ET, Monto AS. Effects of Sequential Influenza A(H1N1)pdm09 Vaccination on Antibody Waning. J Infect Dis 2020; 220:12-19. [PMID: 30722022 DOI: 10.1093/infdis/jiz055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Antibody waning following influenza vaccination has been repeatedly evaluated, but waning has rarely been studied in the context of longitudinal vaccination history. METHODS We developed a Bayesian hierarchical model to assess the effects of sequential influenza A(H1N1)pdm09 vaccination on hemagglutination inhibition antibody boosting and waning in a longitudinal cohort of older children and adults from 2011 to 2016, a period during which the A(H1N1)pdm09 vaccine strain did not change. RESULTS Antibody measurements from 2057 serum specimens longitudinally collected from 388 individuals were included. Average postvaccination antibody titers were similar across successive vaccinations, but the rate of antibody waning increased with each vaccination. The antibody half-life was estimated to decrease from 32 months (95% credible interval [CrI], 22-61 months) following first vaccination to 9 months (95% CrI, 7-15 months) following a seventh vaccination. CONCLUSIONS Although the rate of antibody waning increased with successive vaccination, the estimated antibody half-life was longer than a typical influenza season even among the most highly vaccinated. This supports current recommendations for vaccination at the earliest opportunity. Patterns of boosting and waning might be different with the influenza A(H3N2) subtype, which evolves more rapidly and has been most associated with reduced effectiveness following repeat vaccination.
Collapse
Affiliation(s)
- Jon Zelner
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor.,Department of Center for Social Epidemiology and Population Health, University of Michigan School of Public Health, Ann Arbor
| | - Joshua G Petrie
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor
| | - Rob Trangucci
- Department of Statistics, University of Michigan, Ann Arbor
| | - Emily T Martin
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor
| | - Arnold S Monto
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor
| |
Collapse
|
46
|
Quadrivalent Influenza Vaccine-Induced Antibody Response and Influencing Determinants in Patients ≥ 55 Years of Age in the 2018/2019 Season. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16224489. [PMID: 31739554 PMCID: PMC6887788 DOI: 10.3390/ijerph16224489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023]
Abstract
The effects of immunization with subunit inactivated quadrivalent influenza vaccine (QIV) are not generally well assessed in the elderly Polish population. Therefore, this study evaluated vaccine-induced antibody response and its determinants. Methods: Consecutive patients ≥ 55 years old, attending a Primary Care Clinic in Gryfino, Poland, received QIV (A/Michigan/ 45/2015(H1N1)pdm09, A/Singapore/INFIMH-16-0019/2016 (H3N2), B/Colorado/06/2017, B/Phuket/ 3073/2013) between October-December 2018. Hemagglutination inhibition assays measured antibody response to vaccine strains from pre/postvaccination serum samples. Geometric mean titer ratio (GMTR), protection rate (PR) and seroconversion rate (SR) were also calculated. Results: For 108 patients (54.6% males, mean age: 66.7 years) the highest GMTR (61.5-fold) was observed for A/H3N2/, then B/Colorado/06/2017 (10.3-fold), A/H1N1/pdm09 (8.4-fold) and B/Phuket/ 3073/2013 (3.0-fold). Most patients had post-vaccination protection for A/H3N2/ and B/Phuket/3073/ 2013 (64.8% and 70.4%, respectively); lower PRs were observed for A/H1N1/pdm09 (41.8%) and B/Colorado/06/ 2017 (57.4%). The SRs for A/H3N2/, A/H1N1/pdm09, B Victoria and B Yamagata were 64.8%, 38.0%, 46.8%, and 48.2%, respectively. Patients who received QIV vaccination in the previous season presented lower (p < 0.001 and p = 0.03, respectively) response to B Victoria and B Yamagata. Conclusions: QIV was immunogenic against the additional B lineage strain (B Victoria) without significantly compromising the immunogenicity of the other three vaccine strains, therefore, adding a second B lineage strain in QIV could broaden protection against influenza B infection in this age group. As the QIV immunogenicity differed regarding the four antigens, formulation adjustments to increase the antigen concentration of the serotypes that have lower immunogenicity could increase effectiveness. Prior season vaccination was associated with lower antibody response to a new vaccine, although not consistent through the vaccine strains.
Collapse
|
47
|
Kosikova M, Li L, Radvak P, Ye Z, Wan XF, Xie H. Imprinting of Repeated Influenza A/H3 Exposures on Antibody Quantity and Antibody Quality: Implications for Seasonal Vaccine Strain Selection and Vaccine Performance. Clin Infect Dis 2019; 67:1523-1532. [PMID: 29672713 DOI: 10.1093/cid/ciy327] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/12/2018] [Indexed: 01/04/2023] Open
Abstract
Background Reduced seasonal influenza vaccine effectiveness (VE) was observed in individuals who received repeated annual vaccinations. Preexisting influenza antibody levels were also found inversely correlated with postvaccination titers. These reports suggest that preexisting immunity may affect contemporary seasonal vaccine performance. Methods Influenza A/H3 specific cross-reactivity of postvaccination sera from humans with or without preexisting immunity was assessed by hemagglutination inhibition (HAI) assay. Ferret antisera induced by repeated H3 exposures were also subjected to HAI, antibody affinity, and antibody avidity analyses. Results Human postvaccination sera derived from subjects with or without preexisting immunity showed different cross-reactivity against H3 variant viruses. Similarly, the breadth of cross-reactive ferret antibodies induced by repeated H3 exposures was also broadened. Antigenic differences between H3 viruses characterized by ferret antisera became smaller as the number of exposures increased. Although repeated H3 exposures induced "original antigenic sin" phenomena in HAI titers against later exposed viruses, resultant ferret antibodies showed gradually enhanced avidity for different H3/hemagglutinin. Increased antibody avidity was found to be inversely correlated with decreased antigenic differences among H3 viruses characterized. Conclusions Our results suggest that repeated H3 exposures imprinted not only antibody quantity but also antibody quality. The "naive" ferret model currently used for vaccine strain selection does not recapitulate the complexity of human preexisting immunity. Vaccine strains identified hereby may not provide coverage sufficient for those who were frequently infected and/or vaccinated, leading to the reduced VE observed.
Collapse
Affiliation(s)
- Martina Kosikova
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Lei Li
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University
| | - Peter Radvak
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Zhiping Ye
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Xiu-Feng Wan
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University
| | - Hang Xie
- Laboratory of Pediatric and Respiratory Viral Diseases, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
48
|
Chang LJ, Meng Y, Janosczyk H, Landolfi V, Talbot HK. Safety and immunogenicity of high-dose quadrivalent influenza vaccine in adults ≥65 years of age: A phase 3 randomized clinical trial. Vaccine 2019; 37:5825-5834. [PMID: 31431411 DOI: 10.1016/j.vaccine.2019.08.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND A high-dose, split-virion inactivated trivalent influenza vaccine (IIV3-HD; Fluzone® High-Dose, Sanofi Pasteur) is available for adults ≥65 years of age. This study examined the safety and immunogenicity of a quadrivalent high-dose split-virion inactivated influenza vaccine (IIV4-HD). METHODS This was a randomized, modified double-blind, active-controlled, multi-center trial in healthy adults ≥65 years of age. Subjects were randomized in a 4:1:1 ratio to receive a single intramuscular injection of IIV4-HD, the licensed IIV3-HD, or an IIV3-HD containing the alternate B-lineage strain. Hemagglutination inhibition (HAI), seroneutralisation, and anti-neuraminidase antibody titers were measured at baseline and day 28. Solicited reactions were collected for up to 7 days, unsolicited adverse events up to 28 days, and serious adverse events up to 180 days. The primary immunogenicity objective was to demonstrate that IIV4-HD induces HAI geometric mean titers (GMTs) and seroconversion rates that are non-inferior to those induced by IIV3-HD. Secondary objectives were to describe the safety of IIV4-HD and IIV3-HD and to demonstrate that IIV4-HD induces HAI GMTs and seroconversion rates that are superior to those induced by IIV3-HD not containing the same B-lineage strain. RESULTS The study included 2670 adults ≥65 years of age. For all four strains, HAI GMTs and seroconversion rates induced by IIV4-HD were non-inferior to those induced by IIV3-HDs containing the same strains. For both B strains, HAI GMTs and seroconversion rates induced by IIV4-HD were superior to those induced by IIV3-HD not containing the same B-lineage strain. Seroneutralisation and anti-neuraminidase antibody responses, measured in a subset of subjects, were similar. No new safety concerns were identified, and the safety profiles of IIV4-HD and IIV3-HD were similar. CONCLUSIONS Adding a second B strain in IIV4-HD resulted in improved immunogenicity against the added strain without compromising the immunogenicity of the other strains or the vaccine's tolerability. CLINICAL TRIAL REGISTRATION NCT03282240.
Collapse
Affiliation(s)
- Lee-Jah Chang
- Sanofi Pasteur, 1 Discovery Dr, Swiftwater, PA 18370, USA.
| | - Ya Meng
- Sanofi Pasteur, 1 Discovery Dr, Swiftwater, PA 18370, USA
| | | | | | - H Keipp Talbot
- Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | | |
Collapse
|
49
|
Hay JA, Laurie K, White M, Riley S. Characterising antibody kinetics from multiple influenza infection and vaccination events in ferrets. PLoS Comput Biol 2019; 15:e1007294. [PMID: 31425503 PMCID: PMC6715255 DOI: 10.1371/journal.pcbi.1007294] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/29/2019] [Accepted: 07/29/2019] [Indexed: 12/20/2022] Open
Abstract
The strength and breadth of an individual's antibody repertoire is an important predictor of their response to influenza infection or vaccination. Although progress has been made in understanding qualitatively how repeated exposures shape the antibody mediated immune response, quantitative understanding remains limited. We developed a set of mathematical models describing short-term antibody kinetics following influenza infection or vaccination and fit them to haemagglutination inhibition (HI) titres from 5 groups of ferrets which were exposed to different combinations of trivalent inactivated influenza vaccine (TIV with or without adjuvant), A/H3N2 priming inoculation and post-vaccination A/H1N1 inoculation. We fit models with various immunological mechanisms that have been empirically observed but have not previously been included in mathematical models of antibody landscapes, including: titre ceiling effects, antigenic seniority and exposure-type specific cross reactivity. Based on the parameter estimates of the best supported models, we describe a number of key immunological features. We found quantifiable differences in the degree of homologous and cross-reactive antibody boosting elicited by different exposure types. Infection and adjuvanted vaccination generally resulted in strong, broadly reactive responses whereas unadjuvanted vaccination resulted in a weak, narrow response. We found that the order of exposure mattered: priming with A/H3N2 improved subsequent vaccine response, and the second dose of adjuvanted vaccination resulted in substantially greater antibody boosting than the first. Either antigenic seniority or a titre ceiling effect were included in the two best fitting models, suggesting a role for a mechanism describing diminishing antibody boosting with repeated exposures. Although there was considerable uncertainty in our estimates of antibody waning parameters, our results suggest that both short and long term waning were present and would be identifiable with a larger set of experiments. These results highlight the potential use of repeat exposure animal models in revealing short-term, strain-specific immune dynamics of influenza.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/blood
- Computational Biology
- Cross Reactions
- Disease Models, Animal
- Ferrets/immunology
- Humans
- Immunization, Secondary
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Kinetics
- Models, Immunological
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Vaccines, Inactivated/administration & dosage
Collapse
Affiliation(s)
- James A. Hay
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| | - Karen Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Seqirus, 63 Poplar Road, Parkville, Victoria, Australia
| | - Michael White
- Malaria: Parasites and Hosts, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Steven Riley
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Khurana S, Hahn M, Coyle EM, King LR, Lin TL, Treanor J, Sant A, Golding H. Repeat vaccination reduces antibody affinity maturation across different influenza vaccine platforms in humans. Nat Commun 2019; 10:3338. [PMID: 31350391 PMCID: PMC6659679 DOI: 10.1038/s41467-019-11296-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 06/26/2019] [Indexed: 01/09/2023] Open
Abstract
Several vaccines are approved in the United States for seasonal influenza vaccination every year. Here we compare the impact of repeat influenza vaccination on hemagglutination inhibition (HI) titers, antibody binding and affinity maturation to individual hemagglutinin (HA) domains, HA1 and HA2, across vaccine platforms. Fold change in HI and antibody binding to HA1 trends higher for H1N1pdm09 and H3N2 but not against B strains in groups vaccinated with FluBlok compared with FluCelvax and Fluzone. Antibody-affinity maturation occurs against HA1 domain of H1N1pdm09, H3N2 and B following vaccination with all vaccine platforms, but not against H1N1pdm09-HA2. Importantly, prior year vaccination of subjects receiving repeat vaccinations demonstrated reduced antibody-affinity maturation to HA1 of all three influenza virus strains irrespective of the vaccine platform. This study identifies an important impact of repeat vaccination on antibody-affinity maturation following vaccination, which may contribute to lower vaccine effectiveness of seasonal influenza vaccines in humans Here, Khurana et al. report the results of a phase 4 clinical trial with three FDA approved influenza vaccines and show that repeat influenza vaccination results in reduced antibody affinity maturation to hemagglutinin domain 1 irrespective of vaccine platform.
Collapse
Affiliation(s)
- Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA.
| | - Megan Hahn
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - Elizabeth M Coyle
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - Lisa R King
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - Tsai-Lien Lin
- Division of Biostatistics, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| | - John Treanor
- University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Andrea Sant
- University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Hana Golding
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), FDA, Silver Spring, MD, 20993, USA
| |
Collapse
|