1
|
Shuklina M, Stepanova L, Ozhereleva O, Kovaleva A, Vidyaeva I, Korotkov A, Tsybalova L. Inserting CTL Epitopes of the Viral Nucleoprotein to Improve Immunogenicity and Protective Efficacy of Recombinant Protein against Influenza A Virus. BIOLOGY 2024; 13:801. [PMID: 39452110 PMCID: PMC11505154 DOI: 10.3390/biology13100801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Conserved influenza virus proteins, such as the hemagglutinin stem domain (HA2), nucleoprotein (NP), and matrix protein (M), are the main targets in the development of universal influenza vaccines. Previously, we constructed a recombinant vaccine protein Flg-HA2-2-4M2ehs containing the extracellular domain of the M2 protein (M2e) and the aa76-130 sequence of the second HA subunit as target antigens. It demonstrated immunogenicity and broad protection against influenza A viruses after intranasal and parenteral administration. This study shows that CD8+ epitopes of NP, inserted into a flagellin-fused protein carrying M2e and HA2, affect the post-vaccination immune humoral response to virus antigens without reducing protection. No differences were found between the two proteins in their ability to stimulate the formation of follicular Th in the spleen, which may contribute to a long-lasting antigen-specific humoral response. The data obtained on Balb/c mice suggest that the insertion of CTL NP epitopes into the flagellin-fused protein carrying M2e and HA2 reduces the antibody response to M2e and A/H3N2. In C57Bl6 mice, this stimulates the formation of NP-specific CD8+ Tem and virus-specific mono- and multifunctional CD4+ and CD8+ Tem in the spleen and completely protects mice from influenza virus subtypes A/H1N1pdm09 and A/H3N2.
Collapse
Affiliation(s)
- Marina Shuklina
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, 15/17 Prof. Popova Str., St. Petersburg 197376, Russia
| | - Liudmila Stepanova
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, 15/17 Prof. Popova Str., St. Petersburg 197376, Russia
| | | | | | | | | | | |
Collapse
|
2
|
Nguyen THO, Rowntree LC, Chua BY, Thwaites RS, Kedzierska K. Defining the balance between optimal immunity and immunopathology in influenza virus infection. Nat Rev Immunol 2024; 24:720-735. [PMID: 38698083 DOI: 10.1038/s41577-024-01029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/05/2024]
Abstract
Influenza A viruses remain a global threat to human health, with continued pandemic potential. In this Review, we discuss our current understanding of the optimal immune responses that drive recovery from influenza virus infection, highlighting the fine balance between protective immune mechanisms and detrimental immunopathology. We describe the contribution of innate and adaptive immune cells, inflammatory modulators and antibodies to influenza virus-specific immunity, inflammation and immunopathology. We highlight recent human influenza virus challenge studies that advance our understanding of susceptibility to influenza and determinants of symptomatic disease. We also describe studies of influenza virus-specific immunity in high-risk groups following infection and vaccination that inform the design of future vaccines to promote optimal antiviral immunity, particularly in vulnerable populations. Finally, we draw on lessons from the COVID-19 pandemic to refocus our attention to the ever-changing, highly mutable influenza A virus, predicted to cause future global pandemics.
Collapse
Affiliation(s)
- Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
3
|
Mettelman RC, Souquette A, Van de Velde LA, Vegesana K, Allen EK, Kackos CM, Trifkovic S, DeBeauchamp J, Wilson TL, St James DG, Menon SS, Wood T, Jelley L, Webby RJ, Huang QS, Thomas PG. Baseline innate and T cell populations are correlates of protection against symptomatic influenza virus infection independent of serology. Nat Immunol 2023; 24:1511-1526. [PMID: 37592015 PMCID: PMC10566627 DOI: 10.1038/s41590-023-01590-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Evidence suggests that innate and adaptive cellular responses mediate resistance to the influenza virus and confer protection after vaccination. However, few studies have resolved the contribution of cellular responses within the context of preexisting antibody titers. Here, we measured the peripheral immune profiles of 206 vaccinated or unvaccinated adults to determine how baseline variations in the cellular and humoral immune compartments contribute independently or synergistically to the risk of developing symptomatic influenza. Protection correlated with diverse and polyfunctional CD4+ and CD8+ T, circulating T follicular helper, T helper type 17, myeloid dendritic and CD16+ natural killer (NK) cell subsets. Conversely, increased susceptibility was predominantly attributed to nonspecific inflammatory populations, including γδ T cells and activated CD16- NK cells, as well as TNFα+ single-cytokine-producing CD8+ T cells. Multivariate and predictive modeling indicated that cellular subsets (1) work synergistically with humoral immunity to confer protection, (2) improve model performance over demographic and serologic factors alone and (3) comprise the most important predictive covariates. Together, these results demonstrate that preinfection peripheral cell composition improves the prediction of symptomatic influenza susceptibility over vaccination, demographics or serology alone.
Collapse
Affiliation(s)
- Robert C Mettelman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Aisha Souquette
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lee-Ann Van de Velde
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kasi Vegesana
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - E Kaitlynn Allen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christina M Kackos
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sanja Trifkovic
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jennifer DeBeauchamp
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taylor L Wilson
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Microbiology, Immunology and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Deryn G St James
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Microbiology, Immunology and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Smrithi S Menon
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Timothy Wood
- Institute of Environmental Science and Research Limited (ESR), Wallaceville Science Centre, Upper Hutt, New Zealand
| | - Lauren Jelley
- Institute of Environmental Science and Research Limited (ESR), Wallaceville Science Centre, Upper Hutt, New Zealand
| | - Richard J Webby
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Q Sue Huang
- Institute of Environmental Science and Research Limited (ESR), Wallaceville Science Centre, Upper Hutt, New Zealand.
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
4
|
Körber N, Holzmann-Littig C, Wilkens G, Liao BH, Werz ML, Platen L, Cheng CC, Tellenbach M, Kappler V, Lehner V, Mijočević H, Christa C, Assfalg V, Heemann U, Schmaderer C, Protzer U, Braunisch MC, Bauer T, Renders L. Comparable cellular and humoral immunity upon homologous and heterologous COVID-19 vaccination regimens in kidney transplant recipients. Front Immunol 2023; 14:1172477. [PMID: 37063863 PMCID: PMC10102365 DOI: 10.3389/fimmu.2023.1172477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundKidney transplant recipients (KTRs) are at high risk for a severe course of coronavirus disease 2019 (COVID-19); thus, effective vaccination is critical. However, the achievement of protective immunogenicity is hampered by immunosuppressive therapies. We assessed cellular and humoral immunity and breakthrough infection rates in KTRs vaccinated with homologous and heterologous COVID-19 vaccination regimens.MethodWe performed a comparative in-depth analysis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)–specific T-cell responses using multiplex Fluorospot assays and SARS-CoV-2-specific neutralizing antibodies (NAbs) between three-times homologously (n = 18) and heterologously (n = 8) vaccinated KTRs.ResultsWe detected SARS-CoV-2-reactive T cells in 100% of KTRs upon third vaccination, with comparable frequencies, T-cell expression profiles, and relative interferon γ and interleukin 2 production per single cell between homologously and heterologously vaccinated KTRs. SARS-CoV-2-specific NAb positivity rates were significantly higher in heterologously (87.5%) compared to homologously vaccinated (50.0%) KTRs (P < 0.0001), whereas the magnitudes of NAb titers were comparable between both subcohorts after third vaccination. SARS-CoV-2 breakthrough infections occurred in equal numbers in homologously (38.9%) and heterologously (37.5%) vaccinated KTRs with mild-to-moderate courses of COVID-19.ConclusionOur data support a more comprehensive assessment of not only humoral but also cellular SARS-CoV-2-specific immunity in KTRs to provide an in-depth understanding about the COVID-19 vaccine–induced immune response in a transplant setting.
Collapse
Affiliation(s)
- Nina Körber
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- *Correspondence: Nina Körber,
| | - Christopher Holzmann-Littig
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
- Technical University of Munich (TUM) Medical Education Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Gesa Wilkens
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Maia L. Werz
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Louise Platen
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Myriam Tellenbach
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Verena Kappler
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Viktor Lehner
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Hrvoje Mijočević
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Catharina Christa
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Volker Assfalg
- Department of Surgery, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Matthias C. Braunisch
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
| | - Tanja Bauer
- Institute of Virology, Helmholtz Zentrum München, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Lutz Renders
- Department of Nephrology, Technical University of Munich, School of Medicine, Klinikum Rechts der Isar, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| |
Collapse
|
5
|
Flagellin-Fused Protein Targeting M2e and HA2 Induces Innate and T-Cell Responses in Mice of Different Genetic Lines. Vaccines (Basel) 2022; 10:vaccines10122098. [PMID: 36560509 PMCID: PMC9786633 DOI: 10.3390/vaccines10122098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Efficient control of influenza A infection can potentially be achieved through the development of broad-spectrum vaccines. Recombinant proteins incorporating conserved influenza A virus peptides are one of the platforms for the development of cross-protective influenza vaccines. We constructed a recombinant protein Flg-HA2-2-4M2ehs, in which the extracellular domain of the M2 protein (M2e) and the sequence (aa76-130) of the second subunit of HA (HA2) were used as target antigens. In this study, we investigated the ability of the Flg-HA2-2-4M2ehs protein to activate innate immunity and stimulate the formation of T-cell response in mice of different genetic lines after intranasal immunization. Our studies showed that the Flg-HA2-2-4M2ehs protein was manifested in an increase in the relative content of neutrophils, monocytes, and interstitial macrophages, against the backdrop of a decrease in the level of dendritic cells and increased expression in the CD86 marker. In the lungs of BALB/c mice, immunization with the Flg-HA2-2-4M2ehs protein induced the formation of antigen-specific CD4+ and CD8+ effector memory T cells, producing TNF-α. In mice C57Bl/6, the formation of antigen-specific effector CD8+ T cells, predominantly producing IFN-γ+, was demonstrated. The data obtained showed the formation of CD8+ and CD4+ effector memory T cells expressing the CD107a.
Collapse
|
6
|
Janssens Y, Joye J, Waerlop G, Clement F, Leroux-Roels G, Leroux-Roels I. The role of cell-mediated immunity against influenza and its implications for vaccine evaluation. Front Immunol 2022; 13:959379. [PMID: 36052083 PMCID: PMC9424642 DOI: 10.3389/fimmu.2022.959379] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/27/2022] [Indexed: 12/25/2022] Open
Abstract
Influenza vaccines remain the most effective tools to prevent flu and its complications. Trivalent or quadrivalent inactivated influenza vaccines primarily elicit antibodies towards haemagglutinin and neuraminidase. These vaccines fail to induce high protective efficacy, in particular in older adults and immunocompromised individuals and require annual updates to keep up with evolving influenza strains (antigenic drift). Vaccine efficacy declines when there is a mismatch between its content and circulating strains. Current correlates of protection are merely based on serological parameters determined by haemagglutination inhibition or single radial haemolysis assays. However, there is ample evidence showing that these serological correlates of protection can both over- or underestimate the protective efficacy of influenza vaccines. Next-generation universal influenza vaccines that induce cross-reactive cellular immune responses (CD4+ and/or CD8+ T-cell responses) against conserved epitopes may overcome some of the shortcomings of the current inactivated vaccines by eliciting broader protection that lasts for several influenza seasons and potentially enhances pandemic preparedness. Assessment of cellular immune responses in clinical trials that evaluate the immunogenicity of these new generation vaccines is thus of utmost importance. Moreover, studies are needed to examine whether these cross-reactive cellular immune responses can be considered as new or complementary correlates of protection in the evaluation of traditional and next-generation influenza vaccines. An overview of the assays that can be applied to measure cell-mediated immune responses to influenza with their strengths and weaknesses is provided here.
Collapse
Affiliation(s)
- Yorick Janssens
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
| | - Jasper Joye
- Center for Vaccinology (CEVAC), Ghent University Hospital, Ghent, Belgium
| | - Gwenn Waerlop
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
| | - Frédéric Clement
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
- Center for Vaccinology (CEVAC), Ghent University Hospital, Ghent, Belgium
| | - Isabel Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University, Ghent, Belgium
- Center for Vaccinology (CEVAC), Ghent University Hospital, Ghent, Belgium
- *Correspondence: Isabel Leroux-Roels,
| |
Collapse
|
7
|
Perera DJ, Hassan AS, Liu SS, Elahi SM, Gadoury C, Weeratna RD, Gilbert R, Ndao M. A low dose adenovirus vectored vaccine expressing Schistosoma mansoni Cathepsin B protects from intestinal schistosomiasis in mice. EBioMedicine 2022; 80:104036. [PMID: 35500538 PMCID: PMC9065910 DOI: 10.1016/j.ebiom.2022.104036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/30/2022] [Accepted: 04/16/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Schistosomiasis is an underestimated neglected tropical disease which affects over 236.6 million people worldwide. According to the CDC, the impact of this disease is second to only malaria as the most devastating parasitic infection. Affected individuals manifest chronic pathology due to egg granuloma formation, destroying the liver over time. The only FDA approved drug, praziquantel, does not protect individuals from reinfection, highlighting the need for a prophylactic vaccine. Schistosoma mansoni Cathepsin B (SmCB) is a parasitic gut peptidase necessary for helminth growth and maturation and confers protection as a vaccine target for intestinal schistosomiasis. METHODS An SmCB expressing human adenovirus serotype 5 (AdSmCB) was constructed and delivered intramuscularly to female C57BL/6 mice in a heterologous prime and boost vaccine with recombinant protein. Vaccine induced immunity was described and subsequent protection from parasite infection was assessed by analysing parasite burden and liver pathology. FINDINGS Substantially higher humoral and cell-mediated immune responses, consisting of IgG2c, Th1 effectors, and polyfunctional CD4+ T cells, were induced by the heterologous administration of AdSmCB when compared to the other regimens. Though immune responses favoured Th1 immunity, Th2 responses provided by SmCB protein boosts were maintained. This mixed Th1/Th2 immune response resulted in significant protection from S. mansoni infection comparable to other vaccine formulations which are in clinical trials. Schistosomiasis associated liver pathology was also prevented in a murine model. INTERPRETATION Our study provides missing preclinical data supporting the use of adenoviral vectoring in vaccines for S. mansoni infection. Our vaccination method significantly reduces parasite burden and its associated liver pathology - both of which are critical considerations for this helminth vaccine. FUNDING This work was supported by the Canadian Institutes of Health Research, R. Howard Webster Foundation, and the Foundation of the McGill University Health Centre.
Collapse
Affiliation(s)
- Dilhan J Perera
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Room: EM3.3244, 1001 Decarie Blvd, Montréal, Québec H4A 3J1, Canada
| | - Adam S Hassan
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Room: EM3.3244, 1001 Decarie Blvd, Montréal, Québec H4A 3J1, Canada; Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Sunny S Liu
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | | | | | | | - Rénald Gilbert
- National Research Council Canada, Montréal, Québec, Canada
| | - Momar Ndao
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Room: EM3.3244, 1001 Decarie Blvd, Montréal, Québec H4A 3J1, Canada; Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada; National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
8
|
Sattler A, Schrezenmeier E, Weber UA, Potekhin A, Bachmann F, Straub-Hohenbleicher H, Budde K, Storz E, Proß V, Bergmann Y, Thole LM, Tizian C, Hölsken O, Diefenbach A, Schrezenmeier H, Jahrsdörfer B, Zemojtel T, Jechow K, Conrad C, Lukassen S, Stauch D, Lachmann N, Choi M, Halleck F, Kotsch K. Impaired humoral and cellular immunity after SARS-CoV-2 BNT162b2 (tozinameran) prime-boost vaccination in kidney transplant recipients. J Clin Invest 2021; 131:150175. [PMID: 34101623 PMCID: PMC8279581 DOI: 10.1172/jci150175] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Novel mRNA-based vaccines have been proven to be powerful tools in combating the global pandemic caused by SARS-CoV-2, with BNT162b2 (trade name: Comirnaty) efficiently protecting individuals from COVID-19 across a broad age range. Still, it remains largely unknown how renal insufficiency and immunosuppressive medication affect development of vaccine-induced immunity. We therefore comprehensively analyzed humoral and cellular responses in kidney transplant recipients after the standard second vaccination dose. As opposed to all healthy vaccinees and the majority of hemodialysis patients, only 4 of 39 and 1 of 39 transplanted individuals showed IgA and IgG seroconversion at day 8 ± 1 after booster immunization, with minor changes until day 23 ± 5, respectively. Although most transplanted patients mounted spike-specific T helper cell responses, frequencies were significantly reduced compared with those in controls and dialysis patients and this was accompanied by a broad impairment in effector cytokine production, memory differentiation, and activation-related signatures. Spike-specific CD8+ T cell responses were less abundant than their CD4+ counterparts in healthy controls and hemodialysis patients and almost undetectable in transplant patients. Promotion of anti-HLA antibodies or acute rejection was not detected after vaccination. In summary, our data strongly suggest revised vaccination approaches in immunosuppressed patients, including individual immune monitoring for protection of this vulnerable group at risk of developing severe COVID-19.
Collapse
Affiliation(s)
| | - Eva Schrezenmeier
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrike A. Weber
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Alexander Potekhin
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- MVZ Diaverum Neubrandenburg, Neubrandenburg, Germany
| | - Friederike Bachmann
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Henriette Straub-Hohenbleicher
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Elena Storz
- Department for General and Visceral Surgery and
| | | | | | | | - Caroline Tizian
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Oliver Hölsken
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
- Heidelberg Bioscience International Graduate School, Heidelberg University, Heidelberg, Germany
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Hubert Schrezenmeier
- Department of Transfusion Medicine and Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service, Baden-Württemberg—Hessen and University Hospital Ulm, Ulm University, Ulm, Germany
| | - Bernd Jahrsdörfer
- Department of Transfusion Medicine and Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service, Baden-Württemberg—Hessen and University Hospital Ulm, Ulm University, Ulm, Germany
| | | | | | | | | | - Diana Stauch
- HLA Laboratory, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Nils Lachmann
- HLA Laboratory, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and BIH, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Intensive Care, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | | |
Collapse
|
9
|
Paramsothy A, Lartey Jalloh S, Davies RA, Guttormsen AB, Cox RJ, Mohn KGI. Humoral and cellular immune responses in critically ill influenza A/H1N1-infected patients. Scand J Immunol 2021; 94:e13045. [PMID: 33891354 DOI: 10.1111/sji.13045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/10/2021] [Accepted: 04/11/2021] [Indexed: 12/31/2022]
Abstract
There is limited knowledge of influenza-specific immune responses and their kinetics in critically ill patients. We investigated humoral and cellular immune responses after critical influenza A/H1N1 infection and hypothesized that dysfunctionality or absence of immune responses could contribute to more severe illness. We followed 12 patients hospitalized with severe influenza infection; the majority admitted to intensive care unit (ICU). Blood samples were collected at days 10 and 19 and at 5 months. Antibody responses to surface glycoproteins haemagglutinin (HA) and neuraminidase (NA) of A/H1N1pdm09 were quantified by haemagglutination inhibition (HAI), microneutralization (MN), Enzyme-linked immunosorbent assay (ELISA) and Enzyme-linked lectin assay (ELLA). Influenza-specific antibody levels and avidity were measured separately for head and stalk domains of H1. Cytokine secreting CD4+ and CD8+ T cell responses to conserved influenza epitopes (M1, NP and PB1) were analysed by FluoroSpot. Overall, the patients retained a high level of functional HA- and NA-specific antibodies over the study period. During the acute phase (up to 3 weeks from symptom onset), antibodies specific to H1 stalk increased earlier and were present in higher amount compared with H1 head-specific antibodies. The NA-specific antibodies and the non-neutralizing HA-specific antibody response for H1 head and H1 full-length showed a significant decline from acute to convalescent phase. Despite high total IgG concentrations, avidity to H1 head and H1 full-length protein remained low at all time points. Similarly, CD8+ T cell responses were continuously measured at low levels. In conclusion, our study found that critically ill patients were characterized by low HA-specific antibody avidity and CD8+ T cell response.
Collapse
Affiliation(s)
- Abira Paramsothy
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway
| | - Sarah Lartey Jalloh
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway
| | - Richard A Davies
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway
| | - Anne-Berit Guttormsen
- Department of Anesthesia and Intensive Care, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Rebecca J Cox
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway.,Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Kristin G-I Mohn
- Department of Clinical Sciences, Influenza Centre, University of Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
10
|
Wu CY, Chuang HY, Wong CH. Influenza virus neuraminidase regulates host CD8 + T-cell response in mice. Commun Biol 2020; 3:748. [PMID: 33293641 PMCID: PMC7722854 DOI: 10.1038/s42003-020-01486-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/03/2020] [Indexed: 11/08/2022] Open
Abstract
Influenza A virus (IAV)-specific CD8+ T-cell response was shown to provide protection against pandemic and seasonal influenza infections. However, the response was often relatively weak and the mechanism was unclear. Here, we show that the composition of IAV released from infected cells is regulated by the neuraminidase (NA) activity and the cells infected by NA-defective virus cause intracellular viral protein accumulation and cell death. In addition, after uptake of NA-defective viruses by dendritic cells (DCs), an expression of the major histocompatibility complex class I is induced to activate IAV-specific CD8+ T-cell response. When mice were infected by NA-defective IAV, a CD8+ T-cell response to the highly conserved viral antigens including PB1, NP, HA, M1, M2 and NS1 was observed along with the increasing expression of IL10, IL12 and IL27. Vaccination of mice with NA-defective H1N1 A/WSN/33 induced a strong IAV-specific CD8+ T cell response against H1N1, H3N2 and H5N1. This study reveals the role of NA in the IAV-specific CD8+ T-cell response and virion assembly process, and provides an alternative direction toward the development of universal influenza vaccines.
Collapse
Affiliation(s)
- Chung-Yi Wu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist., Taipei, 115, Taiwan
| | - Hong-Yang Chuang
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist., Taipei, 115, Taiwan
| | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist., Taipei, 115, Taiwan.
- Department of Chemistry, The Scripps Research Institute, 10550N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
11
|
L'huillier AG, Ferreira VH, Hirzel C, Natori Y, Slomovic J, Ku T, Hoschler K, Ierullo M, Selzner N, Schiff J, Singer LG, Humar A, Kumar D. Cell-Mediated Immune Responses After Influenza Vaccination of Solid Organ Transplant Recipients: Secondary Outcomes Analyses of a Randomized Controlled Trial. J Infect Dis 2020; 221:53-62. [PMID: 31550354 DOI: 10.1093/infdis/jiz471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/12/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Despite annual immunization, solid organ transplant (SOT) patients remain at increased risk for severe influenza infection because of suboptimal vaccine immunogenicity. We aimed to compare the CD4+ and CD8+ T-cell responses of the high-dose (HD) and the standard-dose (SD) trivalent inactivated vaccine. METHODS We collected peripheral blood mononuclear cells pre- and postimmunization from 60 patients enrolled in a randomized trial of HD versus SD vaccine (30 HD; 30 SD) during the 2016-2017 influenza season. RESULTS The HD vaccine elicited significantly greater monofunctional and polyfunctional CD4+ and CD8+ T-cell responses against influenza A/H1N1, A/H3N2, and B. For example, median vaccine-elicited influenza-specific polyfunctional CD4+ T cells were higher in recipients of the HD than SD vaccine after stimulation with influenza A/H1N1 (1193 vs 0 per 106 CD4+ T cells; P = .003), A/H3N2 (1154 vs 51; P = .008), and B (1102 vs 0; P = .001). Likewise, vaccine-elicited influenza-specific polyfunctional CD8+ T cells were higher in recipients of the HD than SD vaccine after stimulation with influenza B (367 vs 0; P = .002). CONCLUSIONS Our study provides novel evidence that HD vaccine elicits greater cellular responses compared with the SD vaccine in SOT recipients, which provides support to preferentially consider use of HD vaccination in the SOT setting.
Collapse
Affiliation(s)
| | - Victor H Ferreira
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Cedric Hirzel
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Yoichiro Natori
- Division of Infectious Disease, Department of Medicine, University of Miami Miller School of Medicine and Miami Transplant Institute, Miami, Florida, USA
| | - Jaclyn Slomovic
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Terrance Ku
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | | | - Matthew Ierullo
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Nazia Selzner
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Jeffrey Schiff
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Lianne G Singer
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Atul Humar
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Deepali Kumar
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| |
Collapse
|
12
|
L'Huillier AG, Ferreira VH, Hirzel C, Nellimarla S, Ku T, Natori Y, Humar A, Kumar D. T-cell responses following Natural Influenza Infection or Vaccination in Solid Organ Transplant Recipients. Sci Rep 2020; 10:10104. [PMID: 32572168 PMCID: PMC7308384 DOI: 10.1038/s41598-020-67172-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/11/2020] [Indexed: 11/09/2022] Open
Abstract
Little is known about cell-mediated immune responses to natural influenza infection in solid organ transplant (SOT) patients. The aim of our study was to evaluate the CD4+ and CD8+ responses to influenza A and B infection in a cohort of SOT patients. We collected peripheral blood mononuclear cells at influenza diagnosis and four weeks later from 31 SOT patients during the 2017–2018 influenza season. Infection-elicited influenza-specific CD4+ and CD8+ T-cell responses were measured using flow cytometry and intracellular cytokine staining and compared to responses following influenza vaccine in SOT patients. Natural infection was associated with a significant increase in CD4+ T-cell responses. For example, polyfunctional cells increased from 21 to 782 and from 193 to 1436 cells per 106 CD4+ T-cells among influenza A/H3N2 and B-infected patients (p = 0.006 and 0.004 respectively). Moreover, infection-elicited CD4+ responses were superior than vaccine-elicited responses for influenza A/H1N1 (931 vs 1; p = 0.026), A/H3N2 (647 vs 1; p = 0.041) and B (619 vs 1; p = 0.004). Natural influenza infection triggers a significant increase in CD4+ T-cell responses in SOT patients. Infection elicits significantly stronger CD4+ responses compared to the influenza vaccine and thereby likely elicits better protection against reinfection.
Collapse
Affiliation(s)
- Arnaud G L'Huillier
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University Hospitals of Geneva & University of Geneva Medical School, Geneva, Switzerland.,Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Victor H Ferreira
- Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Cedric Hirzel
- Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada.,Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Terrance Ku
- Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Yoichiro Natori
- Division of Infectious Disease, University of Miami Miller School of Medicine, Miami, Florida, USA.,Miami Transplant Institute, Miami, Florida, USA
| | - Atul Humar
- Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada.
| | - Deepali Kumar
- Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
13
|
Robertson AH, Mahic M, Savic M, Tunheim G, Hungnes O, Trogstad L, Lipkin WI, Mjaaland S. Detection of anti-NS1 antibodies after pandemic influenza exposure: Evaluation of a serological method for distinguishing H1N1pdm09 infected from vaccinated cases. Influenza Other Respir Viruses 2020; 14:294-301. [PMID: 31955522 PMCID: PMC7182603 DOI: 10.1111/irv.12712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/05/2019] [Accepted: 12/08/2019] [Indexed: 12/26/2022] Open
Abstract
Background Reliable exposure information is crucial for assessing health outcomes of influenza infection and vaccination. Current serological methods are unable to distinguish between anti‐hemagglutinin (HA) antibodies induced by infection or vaccination. Objectives We aimed to explore an alternative method for differentiating influenza infection and vaccination. Methods Sera from animals inoculated with influenza viruses or purified H1N1pdm09 HA were obtained. Human samples were selected from a pregnancy cohort established during the 2009 H1N1 pandemic. Unvaccinated, laboratory‐confirmed cases (N = 18), vaccinated cases without influenza‐like‐illness (N = 18) and uninfected, unvaccinated controls (N = 18) were identified based on exposure data from questionnaires, national registries and maternal hemagglutination inhibition (HI) titres at delivery. Animal and human samples were tested for antibodies against the non‐structural protein 1 (NS1) and HA from H1N1pdm09, using a Luciferase Immunoprecipitation System (LIPS). Results Anti‐NS1 H1N1pdm09 antibodies were detected in sera from experimentally infected, but not from vaccinated, animals. Anti‐HA H1N1pdm09 antibodies were detectable after either of these exposures. In human samples, 28% of individuals with laboratory‐confirmed influenza were seropositive for H1N1pdm09 NS1, whereas vaccinated cases and controls were seronegative. There was a trend for H1N1pdm09 NS1 seropositive cases reporting more severe and longer duration of symptomatic illness than seronegative cases. Anti‐HA H1N1pdm09 antibodies were detected in all cases and in 61% of controls. Conclusions The LIPS method could differentiate between sera from experimentally infected and vaccinated animals. However, in human samples obtained more than 6 months after the pandemic, LIPS was specific, but not sufficiently sensitive for ascertaining cases by exposure.
Collapse
Affiliation(s)
- Anna Hayman Robertson
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Milada Mahic
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.,Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Miloje Savic
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Gro Tunheim
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.,KG Jebsen Center for Influenza Vaccine Research, Oslo, Norway
| | - Olav Hungnes
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.,WHO National Influenza Centre, Oslo, Norway
| | - Lill Trogstad
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Walter Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Siri Mjaaland
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.,Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA.,KG Jebsen Center for Influenza Vaccine Research, Oslo, Norway
| |
Collapse
|
14
|
Valkenburg SA, Fang VJ, Leung NHL, Chu DKW, Ip DKM, Perera RAPM, Wang Y, Li APY, Peiris JSM, Cowling BJ, Poon LLM. Cross-reactive antibody-dependent cellular cytotoxicity antibodies are increased by recent infection in a household study of influenza transmission. Clin Transl Immunology 2019; 8:e1092. [PMID: 31763042 PMCID: PMC6864499 DOI: 10.1002/cti2.1092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Influenza causes a spectrum of disease from asymptomatic infection to fatal outcome, and pre-existing immunity can alter susceptibility and disease severity. In a household transmission study, we recruited outpatients with confirmed influenza virus infection and prospectively identified secondary infections in their household contacts, therefore identifying infection cases with baseline samples for determining immune-mediated protection from influenza infection. METHODS We examined baseline broadly reactive immune correlates of relevance to universal vaccine development, specifically antibody-dependent cytotoxic (ADCC) antibodies and T-cell responses in functional assays. Antibodies were assessed in a cell-based NK cell degranulation assay by flow cytometry, and T-cell responses were assessed by IFN-γ intracellular cytokine staining flow cytometry assay. RESULTS The magnitude of antibody responses and ADCC function for multiple influenza-specific proteins was lower in participants who became infected, consolidating the role of pre-existing antibodies in protection from seasonal influenza virus infection. Among H1N1-infected contacts, we found that higher levels of pre-existing H1-haemagglutinin ADCC responses correlated with reduced symptom severity. Recent infection boosted the titre and magnitude of haemagglutinin-, neuraminidase- and nucleoprotein-specific ADCC antibodies. Limited T-cell samples precluded conclusions on the role of pre-existing T-cell responses. CONCLUSIONS Overall, ADCC responses are a protective correlate against influenza virus infection that should be considered in future vaccine development and evaluation.Influenza-specific ADCC responses are elevated in uninfected subjects, associated with reduced symptoms and boosted by recent infection, whilst HA stem and NA IgG are also elevated in uninfected participants irrespective of ADCC function.
Collapse
Affiliation(s)
- Sophie A Valkenburg
- Li Ka Shing Faculty of MedicineHKU Pasteur Research PoleSchool of Public HealthThe University of Hong KongHong Kong
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Vicky J Fang
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Nancy HL Leung
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Daniel KW Chu
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Dennis KM Ip
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Ranawaka APM Perera
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Yizhuo Wang
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Athena PY Li
- Li Ka Shing Faculty of MedicineHKU Pasteur Research PoleSchool of Public HealthThe University of Hong KongHong Kong
| | - JS Malik Peiris
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Benjamin J Cowling
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| | - Leo LM Poon
- Li Ka Shing Faculty of MedicineSchool of Public HealthWHO Collaborating Centre for Infectious Disease Epidemiology and ControlThe University of Hong KongHong Kong
| |
Collapse
|
15
|
Tunheim G, Laake I, Robertson AH, Waalen K, Hungnes O, Naess LM, Cox RJ, Mjaaland S, Trogstad L. Antibody levels in a cohort of pregnant women after the 2009 influenza A(H1N1) pandemic: Waning and association with self-reported severity and duration of illness. Influenza Other Respir Viruses 2018; 13:191-200. [PMID: 30536590 PMCID: PMC6379636 DOI: 10.1111/irv.12623] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/05/2018] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND A population-based pregnancy cohort was established in Norway to study potential effects of exposure to the 2009 influenza pandemic or pandemic vaccination during pregnancy. OBJECTIVES We studied maternal A(H1N1)pdm09-specific hemagglutination inhibition (HI)-titer levels and waning in women with influenza-like illness (ILI) in pregnancy compared to vaccinated women. Moreover, we studied the association between HI-titers and self-reported severity and duration of ILI. METHODS HI-titers against the pandemic virus were measured in maternal blood samples obtained at birth, 3-9 months after exposure, and linked with information about pregnancy, influenza and vaccination from national registries and a cohort questionnaire. RESULTS Among 1821 pregnant women included, 43.7% were unvaccinated and 19.3% of these had ILI. HI-titers were low (geometric mean titer (GMT) 11.3) in the unvaccinated women with ILI. Higher HI-titers (GMT 37.8) were measured in the vaccinated women. Estimated HI-titer waning was similar for vaccinated women and women with ILI. Most ILI episodes were moderate and lasted 3-5 days. Women with ILI reporting specific influenza symptoms such as fever or cough had higher HI-titers than women without these symptoms. Women who reported being "very ill" or illness duration of >5 days had higher HI-titers than women reporting less severe illness or illness of shorter duration, respectively. CONCLUSIONS Antibody waning was similar in vaccinated women and women with ILI. More severe ILI or longer duration of illness was associated with higher HI-titers. Most unvaccinated pregnant women with ILI had low HI-titers, probably due to moderate illness and HI-titer waning between exposure and sampling.
Collapse
Affiliation(s)
- Gro Tunheim
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.,K. G. Jebsen Centre for Influenza Vaccine Research, University of Oslo, Oslo, Norway
| | - Ida Laake
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Anna Hayman Robertson
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Kristian Waalen
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Olav Hungnes
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Lisbeth M Naess
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Rebecca J Cox
- K. G. Jebsen Centre for Influenza Vaccine Research, University of Oslo, Oslo, Norway.,The Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| | - Siri Mjaaland
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.,K. G. Jebsen Centre for Influenza Vaccine Research, University of Oslo, Oslo, Norway
| | - Lill Trogstad
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
16
|
Martin JM, Avula R, Nowalk MP, Lin CJ, Horne WT, Chandran UR, Nagg JP, Zimmerman RK, Cole KS, Alcorn JF. Inflammatory Mediator Expression Associated With Antibody Response Induced by Live Attenuated vs Inactivated Influenza Virus Vaccine in Children. Open Forum Infect Dis 2018; 5:ofy277. [PMID: 30515427 PMCID: PMC6262113 DOI: 10.1093/ofid/ofy277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/23/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The reasons for differences in vaccine effectiveness between live attenuated influenza vaccine (LAIV) and inactivated influenza vaccine (IIV) are not clear. METHODS Blood samples were obtained before vaccination and at days 7 and 21 postvaccination with 2015-2016 quadrivalent IIV or LAIV. Serologic response to the vaccine was measured by hemagglutination inhibition assay. Targeted RNA sequencing and serum cytokine analysis were performed. Paired analyses were used to determine gene expression and were compared between IIV and LAIV recipients. Classification And Regression Trees analysis (CART) identified the strongest associations with vaccine response. RESULTS Forty-six enrollees received IIV, and 25 received LAIV. The mean age was 11.5 (±3.7) years. Seroconversion with IIV was associated with changes in expression of PRKRA and IFI6. Nonseroconversion for both IIV and LAIV was characterized by increased interferon-stimulated gene expression. Seroprotection with both vaccines was associated with altered expression of CXCL2 and CD36. For LAIV, CART showed that changes in expression of CD80, CXCL2, and CASP1 were associated with seroprotection. Serum cytokines showed that IIV seroconversion was associated with decreased CCL3. LAIV seroprotection tracked with decreased tumor necrosis factor-α and interferon-γ. CONCLUSIONS Distinct markers of seroconversion and seroprotection against IIV and LAIV were identified using immunophenotyping and CART analysis.
Collapse
Affiliation(s)
- Judith M Martin
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Raghunandan Avula
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary Patricia Nowalk
- Department of Family Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Chyongchiou Jeng Lin
- Department of Family Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William T Horne
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Uma R Chandran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer P Nagg
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Richard K Zimmerman
- Department of Family Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kelly S Cole
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John F Alcorn
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
17
|
Valkenburg SA, Leung NHL, Bull MB, Yan LM, Li APY, Poon LLM, Cowling BJ. The Hurdles From Bench to Bedside in the Realization and Implementation of a Universal Influenza Vaccine. Front Immunol 2018; 9:1479. [PMID: 30013557 PMCID: PMC6036122 DOI: 10.3389/fimmu.2018.01479] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/14/2018] [Indexed: 12/23/2022] Open
Abstract
Influenza viruses circulate worldwide causing annual epidemics that have a substantial impact on public health. This is despite vaccines being in use for over 70 years and currently being administered to around 500 million people each year. Improvements in vaccine design are needed to increase the strength, breadth, and duration of immunity against diverse strains that circulate during regular epidemics, occasional pandemics, and from animal reservoirs. Universal vaccine strategies that target more conserved regions of the virus, such as the hemagglutinin (HA)-stalk, or recruit other cellular responses, such as T cells and NK cells, have the potential to provide broader immunity. Many pre-pandemic vaccines in clinical development do not utilize new vaccine platforms but use "tried and true" recombinant HA protein or inactivated virus strategies despite substantial leaps in fundamental research on universal vaccines. Significant hurdles exist for universal vaccine development from bench to bedside, so that promising preclinical data is not yet translating to human clinical trials. Few studies have assessed immune correlates derived from asymptomatic influenza virus infections, due to the scale of a study required to identity these cases. The realization and implementation of a universal influenza vaccine requires identification and standardization of set points of protective immune correlates, and consideration of dosage schedule to maximize vaccine uptake.
Collapse
Affiliation(s)
- Sophie A. Valkenburg
- HKU Pasteur Research Pole, The University of Hong Kong, Pokfulam, Hong Kong
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Nancy H. L. Leung
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Maireid B. Bull
- HKU Pasteur Research Pole, The University of Hong Kong, Pokfulam, Hong Kong
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Li-meng Yan
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Athena P. Y. Li
- HKU Pasteur Research Pole, The University of Hong Kong, Pokfulam, Hong Kong
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Leo L. M. Poon
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Benjamin J. Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|