1
|
Ma Y, Dong C, Kim JK, Zhu W, Wei L, Wang Y, Kang SM, Wang BZ. Impact of influenza immune imprinting on immune responses to subsequent vaccinations in mice. Vaccine 2025; 46:126670. [PMID: 39731808 DOI: 10.1016/j.vaccine.2024.126670] [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: 08/14/2024] [Revised: 09/23/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
The immune memory imprinted during an individual's initial influenza exposure (influenza imprinting) has long-lasting effects on the host's response to subsequent influenza infections and vaccinations. Here, we investigate how different influenza virus imprinting impacts the immune responses to subunit, inactivated virus, and protein-based nanoparticle vaccines in Balb/c mice. Our results indicated a phylogenetic distance-dependent effect of influenza imprinting on subunit hemagglutinin (HA) or formalin-inactivated (FI) virus vaccine immunizations. Aichi (H3N2, group 2) HA (HA3) or FI-Aichi vaccination in mice imprinted with closely related Phili (H3N2) triggered significant Aichi-specific HAI antibody and balanced HA3-specific Th1/Th2 antibody immune responses, resulting in robust protection against Aichi. In contrast, HA3 vaccination in PR8 (H1N1, group 1) imprinted mice (PR8-2HA3) induced Th2-leaning responses comparable to those observed in mice without prior influenza immune imprinting (PBS-2HA3). However, subsequent heterosubtypic infections and vaccinations eliminated such effects on antibody subtype profiles. Nonetheless, initial virus exposure established a long-lasting capacity to produce HAI antibody responses against the imprinting strains. Moreover, Phili imprinting followed by HA3/NP nanocluster vaccination protected mice from Aichi infections and induced enhanced cross-reactive immunity. Our study highlights the significance of considering an individual's influenza exposure history when designing and evaluating the effectiveness of influenza vaccines.
Collapse
Affiliation(s)
- Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
2
|
King SM, Bryan SP, Hilchey SP, Wang J, Zand MS. First Impressions Matter: Immune Imprinting and Antibody Cross-Reactivity in Influenza and SARS-CoV-2. Pathogens 2023; 12:169. [PMID: 36839441 PMCID: PMC9967769 DOI: 10.3390/pathogens12020169] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/24/2023] Open
Abstract
Many rigorous studies have shown that early childhood infections leave a lasting imprint on the immune system. The understanding of this phenomenon has expanded significantly since 1960, when Dr. Thomas Francis Jr first coined the term "original antigenic sin", to account for all previous pathogen exposures, rather than only the first. Now more commonly referred to as "immune imprinting", this effect most often focuses on how memory B-cell responses are shaped by prior antigen exposure, and the resultant antibodies produced after subsequent exposure to antigenically similar pathogens. Although imprinting was originally observed within the context of influenza viral infection, it has since been applied to the pandemic coronavirus SARS-CoV-2. To fully comprehend how imprinting affects the evolution of antibody responses, it is necessary to compare responses elicited by pathogenic strains that are both antigenically similar and dissimilar to strains encountered previously. To accomplish this, we must be able to measure the antigenic distance between strains, which can be easily accomplished using data from multidimensional immunological assays. The knowledge of imprinting, combined with antigenic distance measures, may allow for improvements in vaccine design and development for both influenza and SARS-CoV-2 viruses.
Collapse
Affiliation(s)
- Samantha M. King
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Shane P. Bryan
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Shannon P. Hilchey
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jiong Wang
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Martin S. Zand
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, NY 14618, USA
| |
Collapse
|
3
|
Prada LSD, Sanz-Muñoz I, de Lejarazu RO, Eiros JM, García-Sastre A, Aydillo T. Immunodominance hierarchy after seasonal influenza vaccination. Emerg Microbes Infect 2022; 11:2670-2679. [PMID: 36219456 DOI: 10.1080/22221751.2022.2135460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Current influenza vaccines elicit humoral immune responses against the hemagglutinin (HA) protein of influenza viruses. Different antigenic sites have been identified in the HA head as the main target of hemagglutination inhibition (HAI) antibodies (Sb, Sa, Cb, Ca1 and Ca2). To determine immunodominance (ID) of each site, we performed HAI assays against a panel of mutant viruses, each one lacking one of the classically defined antigenic sites and compared it to wild type (Wt). Agglutinating antibodies were measured before and after vaccination in two different regimens: Quadrivalent Influenza Vaccine (QIV) in young adults; or Adjuvanted Trivalent influenza Vaccine (ATIV) in elderly. Our results showed abs before vaccination were significantly reduced against all antigenic sites in the elderly and only against Sb and Ca2 in young adults compared to the Wt. Humoral response to vaccination was significantly reduced against all viruses compared to the Wt for the ATIV and only against Sb and Ca2 for the QIV. The strongest reduction was observed in all cases against Sb followed by Ca2. We concluded that ID profile was clearly dominated by Sb followed by Ca2. Additionally, the antibody response evolved with age, increasing the response towards less immunodominant epitopes of HA head. Adjuvants can positively influence ID hierarchy broadening responses towards multiple antigenic sites of HA head.
Collapse
Affiliation(s)
- Laura Sánchez-de Prada
- National Influenza Centre of Valladolid, 47010, Spain.,Hospital Clínico Universitario de Valladolid, 47003, Spain
| | | | | | | | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
4
|
Auladell M, Phuong HVM, Mai LTQ, Tseng YY, Carolan L, Wilks S, Thai PQ, Price D, Duong NT, Hang NLK, Thanh LT, Thuong NTH, Huong TTK, Diep NTN, Bich VTN, Khvorov A, Hensen L, Duong TN, Kedzierska K, Anh DD, Wertheim H, Boyd SD, Good-Jacobson KL, Smith D, Barr I, Sullivan S, van Doorn HR, Fox A. Influenza virus infection history shapes antibody responses to influenza vaccination. Nat Med 2022; 28:363-372. [PMID: 35177857 DOI: 10.1038/s41591-022-01690-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
Studies of successive vaccination suggest that immunological memory against past influenza viruses may limit responses to vaccines containing current strains. The impact of memory induced by prior infection is rarely considered and is difficult to ascertain, because infections are often subclinical. This study investigated influenza vaccination among adults from the Ha Nam cohort (Vietnam), who were purposefully selected to include 72 with and 28 without documented influenza A(H3N2) infection during the preceding 9 years (Australian New Zealand Clinical Trials Registry 12621000110886). The primary outcome was the effect of prior influenza A(H3N2) infection on hemagglutinin-inhibiting antibody responses induced by a locally available influenza vaccine administered in November 2016. Baseline and postvaccination sera were titrated against 40 influenza A(H3N2) strains spanning 1968-2018. At each time point (baseline, day 14 and day 280), geometric mean antibody titers against 2008-2018 strains were higher among participants with recent infection (34 (29-40), 187 (154-227) and 86 (72-103)) than among participants without recent infection (19 (17-22), 91 (64-130) and 38 (30-49)). On days 14 and 280, mean titer rises against 2014-2018 strains were 6.1-fold (5.0- to 7.4-fold) and 2.6-fold (2.2- to 3.1-fold) for participants with recent infection versus 4.8-fold (3.5- to 6.7-fold) and 1.9-fold (1.5- to 2.3-fold) for those without. One of 72 vaccinees with recent infection versus 4 of 28 without developed symptomatic A(H3N2) infection in the season after vaccination (P = 0.021). The range of A(H3N2) viruses recognized by vaccine-induced antibodies was associated with the prior infection strain. These results suggest that recall of immunological memory induced by prior infection enhances antibody responses to inactivated influenza vaccine and is important to attain protective antibody titers.
Collapse
Affiliation(s)
- Maria Auladell
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | | | | | - Yeu-Yang Tseng
- 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
| | - Louise Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sam Wilks
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Pham Quang Thai
- National Institute of Hygiene and Epidemiology, Ha Noi, Vietnam
| | - David Price
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia.,Victorian Infectious Diseases Reference Laboratory Epidemiology Unit and The Peter Doherty Institute for Infection and Immunity, University of Melbourne and Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | | | | | - Le Thi Thanh
- National Institute of Hygiene and Epidemiology, Ha Noi, Vietnam
| | - Nguyen Thi Hong Thuong
- Oxford University Clinical Research Unit, Wellcome Africa Asia Programme, National Hospital of Tropical Diseases, Ha Noi, Vietnam
| | - Tran Thi Kieu Huong
- Oxford University Clinical Research Unit, Wellcome Africa Asia Programme, National Hospital of Tropical Diseases, Ha Noi, Vietnam
| | - Nguyen Thi Ngoc Diep
- Oxford University Clinical Research Unit, Wellcome Africa Asia Programme, National Hospital of Tropical Diseases, Ha Noi, Vietnam
| | - Vu Thi Ngoc Bich
- Oxford University Clinical Research Unit, Wellcome Africa Asia Programme, National Hospital of Tropical Diseases, Ha Noi, Vietnam
| | - Arseniy Khvorov
- 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
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Tran Nhu Duong
- National Institute of Hygiene and Epidemiology, Ha Noi, Vietnam
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Dang Duc Anh
- National Institute of Hygiene and Epidemiology, Ha Noi, Vietnam
| | - Heiman Wertheim
- Oxford University Clinical Research Unit, Wellcome Africa Asia Programme, National Hospital of Tropical Diseases, Ha Noi, Vietnam.,Department of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
| | - Scott D Boyd
- Stanford University Medical Centre, Stanford University, Stanford, CA, USA
| | - Kim L Good-Jacobson
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Derek Smith
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, UK
| | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sheena Sullivan
- 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
| | - H Rogier van Doorn
- Oxford University Clinical Research Unit, Wellcome Africa Asia Programme, National Hospital of Tropical Diseases, Ha Noi, Vietnam.,Centre of Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Annette Fox
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia. .,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
|
5
|
Safety and Immunogenicity of M2-Deficient, Single Replication, Live Influenza Vaccine (M2SR) in Adults. Vaccines (Basel) 2021; 9:vaccines9121388. [PMID: 34960134 PMCID: PMC8707871 DOI: 10.3390/vaccines9121388] [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: 10/20/2021] [Revised: 11/09/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022] Open
Abstract
M2SR (M2-deficient single replication) is an investigational live intranasal vaccine that protects against multiple influenza A subtypes in influenza-naïve and previously infected ferrets. We conducted a phase 1, first-in-human, randomized, dose-escalation, placebo-controlled study of M2SR safety and immunogenicity. Adult subjects received a single intranasal administration with either placebo or one of three M2SR dose levels (106, 107 or 108 tissue culture infectious dose (TCID50)) expressing hemagglutinin and neuraminidase from A/Brisbane/10/2007 (H3N2) (24 subjects per group). Subjects were evaluated for virus replication, local and systemic reactions, adverse events (AE), and immune responses post-vaccination. Infectious virus was not detected in nasal swabs from vaccinated subjects. At least one AE (most commonly mild nasal rhinorrhea/congestion) was reported among 29%, 58%, and 83% of M2SR subjects administered a low, medium or high dose, respectively, and among 46% of placebo subjects. No subject had fever or a severe reaction to the vaccine. Influenza-specific serum and mucosal antibody responses and B- and T-cell responses were significantly more frequent among vaccinated subjects vs. placebo recipients. The M2SR vaccine was safe and well tolerated and generated dose-dependent durable serum antibody responses against diverse H3N2 influenza strains. M2SR demonstrated a multi-faceted immune response in seronegative and seropositive subjects.
Collapse
|
6
|
McLeod DV, Wahl LM, Mideo N. Mosaic vaccination: How distributing different vaccines across a population could improve epidemic control. Evol Lett 2021; 5:458-471. [PMID: 34621533 PMCID: PMC8484727 DOI: 10.1002/evl3.252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/27/2021] [Indexed: 01/19/2023] Open
Abstract
Although vaccination has been remarkably effective against some pathogens, for others, rapid antigenic evolution results in vaccination conferring only weak and/or short‐lived protection. Consequently, considerable effort has been invested in developing more evolutionarily robust vaccines, either by targeting highly conserved components of the pathogen (universal vaccines) or by including multiple immunological targets within a single vaccine (multi‐epitope vaccines). An unexplored third possibility is to vaccinate individuals with one of a number of qualitatively different vaccines, creating a “mosaic” of individual immunity in the population. Here we explore whether a mosaic vaccination strategy can deliver superior epidemiological outcomes to “conventional” vaccination, in which all individuals receive the same vaccine. We suppose vaccine doses can be distributed between distinct vaccine “targets” (e.g., different surface proteins against which an immune response can be generated) and/or immunologically distinct variants at these targets (e.g., strains); the pathogen can undergo antigenic evolution at both targets. Using simple mathematical models, here we provide a proof‐of‐concept that mosaic vaccination often outperforms conventional vaccination, leading to fewer infected individuals, improved vaccine efficacy, and lower individual risks over the course of the epidemic.
Collapse
Affiliation(s)
- David V McLeod
- Centre D'Ecologie Fonctionnelle & Evolutive CNRS Montpellier 34090 France
| | - Lindi M Wahl
- Mathematics Western University London ON N6A 5B7 Canada
| | - Nicole Mideo
- Department of Ecology and Evolutionary Biology University of Toronto Toronto ON M5S 3B2 Canada
| |
Collapse
|
7
|
Cui C, Wang S, Lu W, Wang Y, Li J, Qu K, Yang M, Wang L, Yu Y. The adjuvanticity of manganese for microbial vaccines via activating the IRF5 signaling pathway. Biochem Pharmacol 2021; 192:114720. [PMID: 34363796 DOI: 10.1016/j.bcp.2021.114720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 11/18/2022]
Abstract
Manganese (Mn2+) has been reported to activate macrophages and NK cells, and to induce the production of type-I interferons (IFNs) by activating the cGAS-STING pathway. Few studies have been conducted on its adjuvanticity to microbial vaccines, and on the involvement of the interferon regulatory factor (IRF) 5 signaling pathway in the adjuvanticity. In this study, we demonstrated that Mn2+ could facilitate various microbial vaccines to induce enhanced antibody responses, and facilitate the influenza virus vaccine to induce protective immunity against the influenza virus challenge. When formulated in vaccines, Mn2+ could activate murine CD4+ T cells, CD8+ T cells, B cells and DCs, and induce the expression and phosphorylation of TANK-binding kinase 1 (TBK1) and IRF5 in the splenocytes of the immunized mice, resulting in the increased expression of type-I IFNs, TNF-α, B cell-activating factor of the TNF family (BAFF) and B lymphocyte-induced maturation protein-1 (Blimp-1). The induced TBK1 could recruit and bind the IRF5. Furthermore, the Mn2+ induced expression of IRF5 and Blimp-1 was prohibited by a IRF5 interfering oligonucleotide. The data suggest the Mn2+ could be used as a novel type of adjuvants for microbial vaccines, and the activation of IRF5 signaling pathway might involve in the adjuvanticity.
Collapse
Affiliation(s)
- Cuiyun Cui
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China
| | - Shengnan Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China
| | - Wenting Lu
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China
| | - Yangyang Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China
| | - Jianhua Li
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China
| | - Kuo Qu
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China
| | - Ming Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China.
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Jilin, Changchun 130021, China.
| |
Collapse
|
8
|
Hasan A, Al-Ozairi E, Al-Baqsumi Z, Ahmad R, Al-Mulla F. Cellular and Humoral Immune Responses in Covid-19 and Immunotherapeutic Approaches. Immunotargets Ther 2021; 10:63-85. [PMID: 33728277 PMCID: PMC7955763 DOI: 10.2147/itt.s280706] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (Covid-19), caused by the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can range in severity from asymptomatic to severe/critical disease. SARS-CoV-2 uses angiotensin-converting enzyme 2 to infect cells leading to a strong inflammatory response, which is most profound in patients who progress to severe Covid-19. Recent studies have begun to unravel some of the differences in the innate and adaptive immune response to SARS-CoV-2 in patients with different degrees of disease severity. These studies have attributed the severe form of Covid-19 to a dysfunctional innate immune response, such as a delayed and/or deficient type I interferon response, coupled with an exaggerated and/or a dysfunctional adaptive immunity. Differences in T-cell (including CD4+ T-cells, CD8+ T-cells, T follicular helper cells, γδ-T-cells, and regulatory T-cells) and B-cell (transitional cells, double-negative 2 cells, antibody-secreting cells) responses have been identified in patients with severe disease compared to mild cases. Moreover, differences in the kinetic/titer of neutralizing antibody responses have been described in severe disease, which may be confounded by antibody-dependent enhancement. Importantly, the presence of preexisting autoantibodies against type I interferon has been described as a major cause of severe/critical disease. Additionally, priorVaccine and multiple vaccine exposure, trained innate immunity, cross-reactive immunity, and serological immune imprinting may all contribute towards disease severity and outcome. Several therapeutic and preventative approaches have been under intense investigations; these include vaccines (three of which have passed Phase 3 clinical trials), therapeutic antibodies, and immunosuppressants.
Collapse
Affiliation(s)
- Amal Hasan
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Ebaa Al-Ozairi
- Clinical Research Unit, Medical Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
- Department of Medicine, Faculty of Medicine, Jabriya, Kuwait City, Kuwait
| | - Zahraa Al-Baqsumi
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Rasheed Ahmad
- Department of Immunology and Microbiology, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Functional Genomics, Research Division, Dasman Diabetes Institute, Dasman, Kuwait City, Kuwait
| |
Collapse
|
9
|
Nikolich-Žugich J, Bradshaw CM, Uhrlaub JL, Watanabe M. Immunity to acute virus infections with advanced age. Curr Opin Virol 2020; 46:45-58. [PMID: 33160186 DOI: 10.1016/j.coviro.2020.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/14/2022]
Abstract
New infections in general, and new viral infections amongst them, represent a serious challenge to an older organism. This review discusses the age-related alterations in responsiveness to infection from the standpoint of virus:host relationship and the host physiological whole-organism and specific immune response to the virus. Changes with age in the innate and adaptive immune system homeostasis and function are reviewed briefly. This is followed by a review of specific alterations and defects in the response of older organisms (chiefly mice and humans) to acute (particularly emerging and re-emerging) viral infections, with a very brief summary of the response to latent persistent infections. Finally, we provide a brief summary of the perspectives for possible interventions to enhance antiviral immunity.
Collapse
Affiliation(s)
- Janko Nikolich-Žugich
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ 85724, USA.
| | - Christine M Bradshaw
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ 85724, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ 85724, USA
| | - Makiko Watanabe
- Department of Immunobiology and the University of Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ 85724, USA
| |
Collapse
|
10
|
Kostoff RN, Kanduc D, Porter AL, Shoenfeld Y, Calina D, Briggs MB, Spandidos DA, Tsatsakis A. Vaccine- and natural infection-induced mechanisms that could modulate vaccine safety. Toxicol Rep 2020; 7:1448-1458. [PMID: 33110761 PMCID: PMC7581376 DOI: 10.1016/j.toxrep.2020.10.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/17/2020] [Indexed: 12/20/2022] Open
Abstract
A degraded/dysfunctional immune system appears to be the main determinant of serious/fatal reaction to viral infection (for COVID-19, SARS, and influenza alike). There are four major approaches being employed or considered presently to augment or strengthen the immune system, in order to reduce adverse effects of viral exposure. The three approaches that are focused mainly on augmenting the immune system are based on the concept that pandemics/outbreaks can be controlled/prevented while maintaining the immune-degrading lifestyles followed by much of the global population. The fourth approach is based on identifying and introducing measures aimed at strengthening the immune system intrinsically in order to minimize future pandemics/outbreaks. Specifically, the four measures are: 1) restricting exposure to virus; 2) providing reactive/tactical treatments to reduce viral load; 3) developing vaccines to prevent, or at least attenuate, the infection; 4) strengthening the immune system intrinsically, by a) identifying those factors that contribute to degrading the immune system, then eliminating/reducing them as comprehensively, thoroughly, and rapidly as possible, and b) replacing the eliminated factors with immune-strengthening factors. This paper focuses on vaccine safety. A future COVID-19 vaccine appears to be the treatment of choice at the national/international level. Vaccine development has been accelerated to achieve this goal in the relatively near-term, and questions have arisen whether vaccine safety has been/is being/will be compromised in pursuit of a shortened vaccine development time. There are myriad mechanisms related to vaccine-induced, and natural infection-induced, infections that could adversely impact vaccine effectiveness and safety. This paper summarizes many of those mechanisms.
Collapse
Affiliation(s)
- Ronald N. Kostoff
- Research Affiliate, School of Public Policy, Georgia Institute of Technology, Gainesville, VA, 20155, USA
| | - Darja Kanduc
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 70125 Bari, Italy
| | - Alan L. Porter
- School of Public Policy, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Search Technology, Inc., Peachtree Corners, GA, 30092, USA
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer 5265601, Israel
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Sechenov University, Moscow, Russia
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71409, Heraklion, Greece
| | - Aristidis Tsatsakis
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation, Sechenov University, Moscow, Russia
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
11
|
Viboud C, Gostic K, Nelson MI, Price GE, Perofsky A, Sun K, Sequeira Trovão N, Cowling BJ, Epstein SL, Spiro DJ. Beyond clinical trials: Evolutionary and epidemiological considerations for development of a universal influenza vaccine. PLoS Pathog 2020; 16:e1008583. [PMID: 32970783 PMCID: PMC7514029 DOI: 10.1371/journal.ppat.1008583] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The prospect of universal influenza vaccines is generating much interest and research at the intersection of immunology, epidemiology, and viral evolution. While the current focus is on developing a vaccine that elicits a broadly cross-reactive immune response in clinical trials, there are important downstream questions about global deployment of a universal influenza vaccine that should be explored to minimize unintended consequences and maximize benefits. Here, we review and synthesize the questions most relevant to predicting the population benefits of universal influenza vaccines and discuss how existing information could be mined to begin to address these questions. We review three research topics where computational modeling could bring valuable evidence: immune imprinting, viral evolution, and transmission. We address the positive and negative consequences of imprinting, in which early childhood exposure to influenza shapes and limits immune responses to future infections via memory of conserved influenza antigens. However, the mechanisms at play, their effectiveness, breadth of protection, and the ability to "reprogram" already imprinted individuals, remains heavily debated. We describe instances of rapid influenza evolution that illustrate the plasticity of the influenza virus in the face of drug pressure and discuss how novel vaccines could introduce new selective pressures on the evolution of the virus. We examine the possible unintended consequences of broadly protective (but infection-permissive) vaccines on the dynamics of epidemic and pandemic influenza, compared to conventional vaccines that have been shown to provide herd immunity benefits. In conclusion, computational modeling offers a valuable tool to anticipate the benefits of ambitious universal influenza vaccine programs, while balancing the risks from endemic influenza strains and unpredictable pandemic viruses. Moving forward, it will be important to mine the vast amount of data generated in clinical studies of universal influenza vaccines to ensure that the benefits and consequences of these vaccine programs have been carefully modeled and explored.
Collapse
Affiliation(s)
- Cécile Viboud
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States
- * E-mail:
| | - Katelyn Gostic
- Dept. of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, California, United States
- Dept. of Ecology and Evolution, University of Chicago, Chicago, Illinois, United States
| | - Martha I. Nelson
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Graeme E. Price
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Amanda Perofsky
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Kaiyuan Sun
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Nídia Sequeira Trovão
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Benjamin J. Cowling
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Suzanne L. Epstein
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States
| | - David J. Spiro
- Division of International Epidemiology and Population Studies, Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
12
|
Impact of Pre-Existing Immunity on Live Attenuated Influenza Vaccine-Induced Cross-Protective Immunity. Vaccines (Basel) 2020; 8:vaccines8030459. [PMID: 32825218 PMCID: PMC7563680 DOI: 10.3390/vaccines8030459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022] Open
Abstract
The efficacy of the intranasally (i.n.) delivered live attenuated influenza vaccine (LAIV) is variable and, in some seasons, suboptimal. In this study, we report that LAIV exhibits cross-protective efficacy in mice, potentially associated with cellular immunity as opposed to antigen-specific antibody responses. However, pre-exposure to the intramuscularly (i.m.) delivered inactivated influenza vaccine (IIV) severely impaired LAIV-induced cross-protection against heterologous challenge, potentially by inhibiting replication of LAIV. Our findings suggest that pre-existing immunity afforded by IIV suppresses cross-protective T cell immunogenicity of LAIV.
Collapse
|
13
|
Song EJ, Españo E, Nam JH, Kim J, Shim KS, Shin E, Park YI, Lee CK, Kim JK. Adjuvanticity of Processed Aloe vera gel for Influenza Vaccination in Mice. Immune Netw 2020; 20:e31. [PMID: 32895618 PMCID: PMC7458799 DOI: 10.4110/in.2020.20.e31] [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: 06/01/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/01/2022] Open
Abstract
The effectiveness of current influenza vaccines is considered suboptimal, and 1 way to improve the vaccines is using adjuvants. However, the current pool of adjuvants used in influenza vaccination is limited due to safety concerns. Aloe vera, or aloe, has been shown to have immunomodulatory functions and to be safe for oral intake. In this study, we explored the potential of orally administered processed Aloe vera gel (PAG) as an adjuvant for influenza vaccines in C57BL/6 mice. We first evaluated its adjuvanticity with a split-type pandemic H1N1 (pH1N1) Ag by subjecting the mice to lethal homologous influenza challenge. Oral PAG administration with the pH1N1 Ag increased survival rates in mice to levels similar to those of alum and MF59, which are currently used as adjuvants in influenza vaccine formulations. Similarly, oral PAG administration improved the survival of mice immunized with a commercial trivalent influenza vaccine against lethal homologous and heterologous virus challenge. PAG also increased hemagglutination inhibition and virus neutralization Ab titers against homologous and heterologous influenza strains following immunization with the split-type pH1N1 Ag or the commercial trivalent vaccine. Therefore, this study demonstrates that PAG may potentially be used as an adjuvant for influenza vaccines.
Collapse
Affiliation(s)
- Eun-Jung Song
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea.,Department of Veterinary Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea
| | - Erica Españo
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| | - Jeong-Hyun Nam
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea.,Division of Viral Disease Research, Center for Infectious Disease Research, National Institute of Health, Korea Centers for Disease Control and Prevention, Cheongju 28159, Korea
| | - Jiyeon Kim
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| | | | | | - Young In Park
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| | - Chong-Kil Lee
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Jeong-Ki Kim
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| |
Collapse
|
14
|
Dhakal S, Klein SL. Host Factors Impact Vaccine Efficacy: Implications for Seasonal and Universal Influenza Vaccine Programs. J Virol 2019; 93:e00797-19. [PMID: 31391269 PMCID: PMC6803252 DOI: 10.1128/jvi.00797-19] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected. Growing evidences suggest that host factors including age, biological sex, pregnancy, and immune history play important roles as modifiers of influenza virus vaccine efficacy. We hypothesize that host genetics, the hormonal milieu, and gut microbiota contribute to host-related differences in influenza virus vaccine efficacy. This review highlights the current insights and future perspectives into host-specific factors that impact influenza vaccine-induced immunity and protection. Consideration of the host factors that affect influenza vaccine-induced immunity might improve influenza vaccines by providing empirical evidence for optimizing or even personalizing vaccine type, dose, and use of adjuvants for current seasonal and future universal influenza vaccines.
Collapse
Affiliation(s)
- Santosh Dhakal
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Host Factors Impact Vaccine Efficacy: Implications for Seasonal and Universal Influenza Vaccine Programs. J Virol 2019. [PMID: 31391269 DOI: 10.1128/jvi.00797‐19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Influenza is a global public health problem. Current seasonal influenza vaccines have highly variable efficacy, and thus attempts to develop broadly protective universal influenza vaccines with durable protection are under way. While much attention is given to the virus-related factors contributing to inconsistent vaccine responses, host-associated factors are often neglected. Growing evidences suggest that host factors including age, biological sex, pregnancy, and immune history play important roles as modifiers of influenza virus vaccine efficacy. We hypothesize that host genetics, the hormonal milieu, and gut microbiota contribute to host-related differences in influenza virus vaccine efficacy. This review highlights the current insights and future perspectives into host-specific factors that impact influenza vaccine-induced immunity and protection. Consideration of the host factors that affect influenza vaccine-induced immunity might improve influenza vaccines by providing empirical evidence for optimizing or even personalizing vaccine type, dose, and use of adjuvants for current seasonal and future universal influenza vaccines.
Collapse
|
16
|
Hill EM, Petrou S, de Lusignan S, Yonova I, Keeling MJ. Seasonal influenza: Modelling approaches to capture immunity propagation. PLoS Comput Biol 2019; 15:e1007096. [PMID: 31658250 PMCID: PMC6837557 DOI: 10.1371/journal.pcbi.1007096] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 11/07/2019] [Accepted: 10/01/2019] [Indexed: 11/18/2022] Open
Abstract
Seasonal influenza poses serious problems for global public health, being a significant contributor to morbidity and mortality. In England, there has been a long-standing national vaccination programme, with vaccination of at-risk groups and children offering partial protection against infection. Transmission models have been a fundamental component of analysis, informing the efficient use of limited resources. However, these models generally treat each season and each strain circulating within that season in isolation. Here, we amalgamate multiple data sources to calibrate a susceptible-latent-infected-recovered type transmission model for seasonal influenza, incorporating the four main strains and mechanisms linking prior season epidemiological outcomes to immunity at the beginning of the following season. Data pertaining to nine influenza seasons, starting with the 2009/10 season, informed our estimates for epidemiological processes, virological sample positivity, vaccine uptake and efficacy attributes, and general practitioner influenza-like-illness consultations as reported by the Royal College of General Practitioners (RCGP) Research and Surveillance Centre (RSC). We performed parameter inference via approximate Bayesian computation to assess strain transmissibility, dependence of present season influenza immunity on prior protection, and variability in the influenza case ascertainment across seasons. This produced reasonable agreement between model and data on the annual strain composition. Parameter fits indicated that the propagation of immunity from one season to the next is weaker if vaccine derived, compared to natural immunity from infection. Projecting the dynamics forward in time suggests that while historic immunity plays an important role in determining annual strain composition, the variability in vaccine efficacy hampers our ability to make long-term predictions.
Collapse
Affiliation(s)
- Edward M. Hill
- Zeeman Institute: Systems Biology and Infectious Disease Epidemiology Research (SBIDER), University of Warwick, Coventry, United Kingdom
- Mathematics Institute, University of Warwick, Coventry, United Kingdom
| | - Stavros Petrou
- Warwick Clinical Trials Unit, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom
- Royal College of General Practitioners, London, United Kingdom
| | - Ivelina Yonova
- Royal College of General Practitioners, London, United Kingdom
- Department of Clinical and Experimental Medicine, University of Surrey, Guildford, United Kingdom
| | - Matt J. Keeling
- Zeeman Institute: Systems Biology and Infectious Disease Epidemiology Research (SBIDER), University of Warwick, Coventry, United Kingdom
- Mathematics Institute, University of Warwick, Coventry, United Kingdom
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
17
|
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
|
18
|
Fox A, Quinn KM, Subbarao K. Extending the Breadth of Influenza Vaccines: Status and Prospects for a Universal Vaccine. Drugs 2019; 78:1297-1308. [PMID: 30088204 DOI: 10.1007/s40265-018-0958-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite the widespread use of seasonal influenza vaccines, there is urgent need for a universal influenza vaccine to provide broad, long-term protection. A number of factors underpin this urgency, including threats posed by zoonotic and pandemic influenza A viruses, suboptimal effectiveness of seasonal influenza vaccines, and concerns surrounding the effects of annual vaccination. In this article, we discuss approaches that are being investigated to increase influenza vaccine breadth, which are near-term, readily achievable approaches to increase the range of strains recognized within a subtype, or longer-term more challenging approaches to produce a truly universal influenza vaccine. Adjuvanted and neuraminidase-optimized vaccines are emerging as the most feasible and promising approaches to extend protection to cover a broader range of strains within a subtype. The goal of developing a universal vaccine has also been advanced with the design of immunogenic influenza HA-stem constructs that induce broadly neutralizing antibodies. However, these constructs are not yet sufficiently immunogenic to induce lasting universal immunity in humans. Advances in understanding how T cells mediate protection, and how viruses are packaged, have facilitated the rationale design and delivery of replication-incompetent virus vaccines that induce broad protection mediated by lung-resident memory T cells. While the lack of clear mechanistic correlates of protection, other than haemagglutination-inhibiting antibodies, remains an impediment to further advancing novel influenza vaccines, the pressing need for such a vaccine is supporting development of highly innovative and effective strategies.
Collapse
Affiliation(s)
- Annette Fox
- WHO Collaborating Centre for Reference and Research on Influenza, and the Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC, Australia
| | - Kylie M Quinn
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, and the Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC, Australia.
| |
Collapse
|
19
|
Li Z, Song Y, Cui C, Lan Y, Li X, Liu Y, Lu F, Zhang Y, Yu Y, Wang L. A LAG3-interfering oligonucleotide acts as an adjuvant to enhance the antibody responses induced by recombinant protein vaccines and inactivated influenza virus vaccines. Appl Microbiol Biotechnol 2019; 103:6543-6557. [PMID: 31236615 DOI: 10.1007/s00253-019-09919-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/04/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023]
Abstract
Lymphocyte activation gene-3 (LAG3) is a transmembrane protein expressed on activated T cells and delivers inhibitory signals to render the T cells unable to effectively help B cells to produce antibodies to microbes and vaccines. Presumably, antagonizing LAG3 could enhance the antibody responses to vaccines, and LAG3 antagonists could facilitate vaccines to induce vigorous antibody responses. In this study, we designed a LAG3-interfering antisense oligonucleotide, designated as LIO-1. The LIO-1 is complementary to an identical region shared in human and mouse LAG3 mRNA. We demonstrated that LIO-1 induced the degradation of LAG3 mRNA in immune cells, decreased the LAG3 expression on CD4+ T cells, maintained the prolonged proliferation and promoted the activation of antigen-specific CD4+ T cells, and increased the production of IFN-γ, IL-2, and IL-6 in the antigen re-stimulated immune cells. In addition, we found that LIO-1 enhanced the antibody responses induced by ISA35-formulated recombinant antigen vaccine or ISA35-formulated inactivated influenza virus vaccines in mice. Thus, the LIO-1, a nucleic acid LAG3 antagonist, could facilitate vaccines to induce vigorous antibody responses and has the possibility to be used as a novel adjuvant.
Collapse
Affiliation(s)
- Zhiqin Li
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Yilan Song
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Cuiyun Cui
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Yu Lan
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Xin Li
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Ye Liu
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Fangjie Lu
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Ya Zhang
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China.
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
20
|
Lee J, Paparoditis P, Horton AP, Frühwirth A, McDaniel JR, Jung J, Boutz DR, Hussein DA, Tanno Y, Pappas L, Ippolito GC, Corti D, Lanzavecchia A, Georgiou G. Persistent Antibody Clonotypes Dominate the Serum Response to Influenza over Multiple Years and Repeated Vaccinations. Cell Host Microbe 2019; 25:367-376.e5. [PMID: 30795981 DOI: 10.1016/j.chom.2019.01.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/02/2018] [Accepted: 01/18/2019] [Indexed: 12/16/2022]
Abstract
Humans are repeatedly exposed to influenza virus via infections and vaccinations. Understanding how multiple exposures and pre-existing immunity impact antibody responses is essential for vaccine development. Given the recent prevalence of influenza H1N1 A/California/7/2009 (CA09), we examined the clonal composition and dynamics of CA09 hemagglutinin (HA)-reactive IgG repertoire over 5 years in a donor with multiple influenza exposures. The anti-CA09 HA polyclonal response in this donor comprised 24 persistent antibody clonotypes, accounting for 72.6% ± 10.0% of the anti-CA09 HA repertoire over 5 years. These persistent antibodies displayed higher somatic hypermutation relative to transient serum antibodies detected at one time point. Additionally, persistent antibodies predominantly demonstrated cross-reactivity and potent neutralization toward a phylogenetically distant H5N1 A/Vietnam/1203/2004 (VT04) strain, a feature correlated with HA stem recognition. This analysis reveals how "serological imprinting" impacts responses to influenza and suggests that once elicited, cross-reactive antibodies targeting the HA stem can persist for years.
Collapse
Affiliation(s)
- Jiwon Lee
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Philipp Paparoditis
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland; Institute of Microbiology, ETH Zürich, Zürich 8093, Switzerland
| | - Andrew P Horton
- Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Alexander Frühwirth
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Jonathan R McDaniel
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiwon Jung
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Daniel R Boutz
- Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Dania A Hussein
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yuri Tanno
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Leontios Pappas
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland
| | - Gregory C Ippolito
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | | | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona 6500, Switzerland; Institute of Microbiology, ETH Zürich, Zürich 8093, Switzerland
| | - George Georgiou
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
21
|
Elicitation of Protective Antibodies against 20 Years of Future H3N2 Cocirculating Influenza Virus Variants in Ferrets Preimmune to Historical H3N2 Influenza Viruses. J Virol 2019; 93:JVI.00946-18. [PMID: 30429350 DOI: 10.1128/jvi.00946-18] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/22/2018] [Indexed: 01/16/2023] Open
Abstract
The vast majority of people already have preexisting immune responses to influenza viruses from one or more subtypes. However, almost all preclinical studies evaluate new influenza vaccine candidates in immunologically naive animals. Recently, our group demonstrated that priming naive ferrets with broadly reactive H1 COBRA HA-based vaccines boosted preexisting antibodies induced by wild-type H1N1 virus infections. These H1 COBRA hemagglutinin (HA) antigens induced antibodies with HAI activity against multiple antigenically different H1N1 viral variants. In this study, ferrets, preimmune to historical H3N2 viruses, were vaccinated with virus-like particle (VLP) vaccines expressing either an HA from a wild-type H3 influenza virus or a COBRA H3 HA antigen (T6, T7, T10, or T11). The elicited antisera had the ability to neutralize virus infection against either a panel of viruses representing vaccine strains selected by the World Health Organization or a set of viral variants that cocirculated during the same time period. Preimmune animals vaccinated with H3 COBRA T10 HA antigen elicited sera with higher hemagglutination inhibition (HAI) antibody titers than antisera elicited by VLP vaccines with wild-type HA VLPs in preimmune ferrets. However, while the T11 COBRA vaccine did not elicit HAI activity, the elicited antibodies did neutralize antigenically distinct H3N2 influenza viruses. Overall, H3 COBRA-based HA vaccines were able to neutralize both historical H3 and contemporary, as well as future, H3N2 viruses with higher titers than vaccines with wild-type H3 HA antigens. This is the first report demonstrating the effectiveness of a broadly reactive H3N3 vaccine in a preimmune ferret model.IMPORTANCE After exposure to influenza virus, the host generates neutralizing anti-hemagglutinin (anti-HA) antibodies against that specific infecting influenza strain. These antibodies can also neutralize some, but not all, cocirculating strains. The goal of next-generation influenza vaccines, such as HA head-based COBRA, is to stimulate broadly protective neutralizing antibodies against all strains circulating within a subtype, in particular those that persist over multiple influenza seasons, without requiring an update to the vaccine. To mimic the human condition, COBRA HA virus-like particle vaccines were tested in ferrets that were previously exposed to historical H3N2 influenza viruses. In this model, these vaccines elicited broadly protective antibodies that neutralized cocirculating H3N2 influenza viruses isolated over a 20-year period. This is the first study to show the effectiveness of H3N3 COBRA HA vaccines in a host with preexisting immunity to influenza.
Collapse
|
22
|
Antibody Responses toward the Major Antigenic Sites of Influenza B Virus Hemagglutinin in Mice, Ferrets, and Humans. J Virol 2019; 93:JVI.01673-18. [PMID: 30381487 DOI: 10.1128/jvi.01673-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022] Open
Abstract
The influenza B virus hemagglutinin contains four major antigenic sites (the 120 loop, the 150 loop, the 160 loop, and the 190 helix) within the head domain. These immunodominant antigenic sites are the main targets of neutralizing antibodies and are subject to antigenic drift. Yet little is known about the specific antibody responses toward each site in terms of antibody prevalence and hemagglutination inhibition activity. In this study, we used modified hemagglutinins of influenza B virus which display only one or none of the major antigenic sites to measure antibody responses toward the classical as well as the noncanonical epitopes in mice, ferrets, and humans. With our novel reagents, we found that both hemagglutination inhibition antibodies and total IgGs were mostly induced by the major antigenic sites. However, in human adults, we observed high hemagglutination inhibition antibody responses toward the noncanonical epitopes. By stratifying the human samples into age groups, we found that the noncanonical antibody responses appeared to increase with age.IMPORTANCE This study dissected the specific antibody responses toward the major antigenic sites and the noncanonical epitopes of influenza B virus hemagglutinin in animals and humans using novel reagents. These findings will guide the design of the next generation of influenza virus vaccines.
Collapse
|
23
|
Gage E, Van Hoeven N, Dubois Cauwelaert N, Larsen SE, Erasmus J, Orr MT, Coler RN. Memory CD4 + T cells enhance B-cell responses to drifting influenza immunization. Eur J Immunol 2018; 49:266-276. [PMID: 30548475 DOI: 10.1002/eji.201847852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/13/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022]
Abstract
Influenza A annually infects 5-10% of the world's human population resulting in one million deaths. Influenza causes annual epidemics and reinfects previously exposed individuals because of antigenic drift in the glycoprotein hemagglutinin. Due to antigenic drift, the immune system is simultaneously exposed to novel and conserved parts of the influenza virus via vaccination and/or infection throughout life. Preexisting immunity has long been known to augment subsequent hemagglutination inhibitory antibody (hAb) responses. However, the preexisting immunological contributors that influence hAb responses are not understood. Therefore, we adapted and developed sequential infection and immunization mouse models using drifted influenza strains to show that MHC Class II haplotype and T-cell reactivity influences subsequent hAb responses. We found that CB6F1 mice infected with A/CA followed by immunization with A/PR8 have increased hAb responses to A/PR8 compared to C57BL/6 mice. Increased hAb responses in CB6F1 mice were CD4+ T-cell and B-cell dependent and corresponded to increased germinal center A/PR8-specific B and T-follicular helper cells. These results suggest conserved MHC Class II restricted epitopes within HA are essential for B cells to respond to drifting influenza and could be leveraged to boost hAb responses.
Collapse
Affiliation(s)
- Emily Gage
- Infectious Disease Research Institute, Seattle, WA, USA.,Department of Global Health, University of Washington, Seattle, WA, USA
| | - Neal Van Hoeven
- Infectious Disease Research Institute, Seattle, WA, USA.,Department of Global Health, University of Washington, Seattle, WA, USA.,PAI Life Sciences, Seattle, WA, USA
| | | | | | - Jesse Erasmus
- Infectious Disease Research Institute, Seattle, WA, USA
| | - Mark T Orr
- Infectious Disease Research Institute, Seattle, WA, USA.,Department of Global Health, University of Washington, Seattle, WA, USA
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA, USA.,Department of Global Health, University of Washington, Seattle, WA, USA.,PAI Life Sciences, Seattle, WA, USA
| |
Collapse
|
24
|
Valenciano M, Kissling E, Larrauri A, Nunes B, Pitigoi D, O'Donnell J, Reuss A, Horváth JK, Paradowska‐Stankiewicz I, Rizzo C, Falchi A, Daviaud I, Brytting M, Meijer A, Kaic B, Gherasim A, Machado A, Ivanciuc A, Domegan L, Schweiger B, Ferenczi A, Korczyńska M, Bella A, Vilcu A, Mosnier A, Zakikhany K, de Lange M, Kurečić Filipovićović S, Johansen K, Moren A. Exploring the effect of previous inactivated influenza vaccination on seasonal influenza vaccine effectiveness against medically attended influenza: Results of the European I-MOVE multicentre test-negative case-control study, 2011/2012-2016/2017. Influenza Other Respir Viruses 2018; 12:567-581. [PMID: 29659149 PMCID: PMC6086844 DOI: 10.1111/irv.12562] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Results of previous influenza vaccination effects on current season influenza vaccine effectiveness (VE) are inconsistent. OBJECTIVES To explore previous influenza vaccination effects on current season VE among population targeted for vaccination. METHODS We used 2011/2012 to 2016/2017 I-MOVE primary care multicentre test-negative data. For each season, we compared current season adjusted VE (aVE) between individuals vaccinated and unvaccinated in previous season. Using unvaccinated in both seasons as a reference, we then compared aVE between vaccinated in both seasons, current only, and previous only. RESULTS We included 941, 2645 and 959 influenza-like illness patients positive for influenza A(H1N1)pdm09, A(H3N2) and B, respectively, and 5532 controls. In 2011/2012, 2014/2015 and 2016/2017, A(H3N2) aVE point estimates among those vaccinated in previous season were -68%, -21% and -19%, respectively; among unvaccinated in previous season, these were 33%, 48% and 46%, respectively (aVE not computable for influenza A(H1N1)pdm09 and B). Compared to current season vaccination only, VE for both seasons' vaccination was (i) similar in two of four seasons for A(H3N2) (absolute difference [ad] 6% and 8%); (ii) lower in three of four seasons for influenza A(H1N1)pdm09 (ad 18%, 26% and 29%), in two seasons for influenza A(H3N2) (ad 27% and 39%) and in two of three seasons for influenza B (ad 26% and 37%); (iii) higher in one season for influenza A(H1N1)pdm09 (ad 20%) and influenza B (ad 24%). CONCLUSIONS We did not identify any pattern of previous influenza vaccination effect. Prospective cohort studies documenting influenza infections, vaccinations and vaccine types are needed to understand previous influenza vaccinations' effects.
Collapse
Affiliation(s)
| | | | - Amparo Larrauri
- National Centre of EpidemiologyInstitute of Health Carlos IIIMadridSpain
| | - Baltazar Nunes
- Department of EpidemiologyInstituto Nacional de Saúde, Doctor Ricardo JorgeLisboaPortugal
| | - Daniela Pitigoi
- University of Medicine and Pharmacy Carol DavilaBucharestRomania
- Cantacuzino InstituteNational Institute of Research – Development for Microbiology and ImmunologyBucharestRomania
| | - Joan O'Donnell
- Health Service Executive – Health Protection Surveillance CentreDublinIreland
| | - Annicka Reuss
- Department for Infectious Disease EpidemiologyRobert Koch InstituteBerlinGermany
| | - Judit Krisztina Horváth
- Department of Disease Prevention and SurveillanceNational Centre for EpidemiologyBudapestHungary
| | | | - Caterina Rizzo
- National Center for Epidemiology, Surveillance and Health PromotionIstituto Superiore di SanitàRomeItaly
| | | | | | - Mia Brytting
- The Public Health Agency of SwedenStockholmSweden
| | - Adam Meijer
- Centre for Infectious Disease ControlNational Institute of Public Health and Environment (RIVM)BilthovenThe Netherlands
| | | | - Alin Gherasim
- National Centre of EpidemiologyInstitute of Health Carlos IIIMadridSpain
| | - Ausenda Machado
- Department of EpidemiologyInstituto Nacional de Saúde, Doctor Ricardo JorgeLisboaPortugal
| | - Alina Ivanciuc
- Cantacuzino InstituteNational Institute of Research – Development for Microbiology and ImmunologyBucharestRomania
| | - Lisa Domegan
- Health Service Executive – Health Protection Surveillance CentreDublinIreland
| | - Brunhilde Schweiger
- Department for Infectious Disease EpidemiologyRobert Koch InstituteBerlinGermany
| | - Annamária Ferenczi
- Department of Disease Prevention and SurveillanceNational Centre for EpidemiologyBudapestHungary
| | - Monika Korczyńska
- National Institute of Public Health – National Institute of HygieneWarsawPoland
| | - Antonino Bella
- National Center for Epidemiology, Surveillance and Health PromotionIstituto Superiore di SanitàRomeItaly
| | - Ana‐Maria Vilcu
- Institut Pierre Louis d'épidémiologie et de Santé Publique (IPLESP UMRS 1136)UPMC Univ Paris 06, INSERMSorbonne UniversitésParisFrance
| | | | | | - Marit de Lange
- Centre for Infectious Disease ControlNational Institute of Public Health and Environment (RIVM)BilthovenThe Netherlands
| | | | - Kari Johansen
- European Centre for Disease Prevention and Control (ECDC)StockholmSweden
| | | | | |
Collapse
|
25
|
Harnessing immune history to combat influenza viruses. Curr Opin Immunol 2018; 53:187-195. [DOI: 10.1016/j.coi.2018.05.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 01/23/2023]
|
26
|
Cobey S, Hensley SE. Immune history and influenza virus susceptibility. Curr Opin Virol 2017; 22:105-111. [PMID: 28088686 DOI: 10.1016/j.coviro.2016.12.004] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/14/2016] [Accepted: 12/20/2016] [Indexed: 12/25/2022]
Abstract
Antibody responses to influenza viruses are critical for protection, but the ways in which repeated viral exposures shape antibody evolution and effectiveness over time remain controversial. Early observations demonstrated that viral exposure history has a profound effect on the specificity and magnitude of antibody responses to a new viral strain, a phenomenon called 'original antigenic sin.' Although 'sin' might suppress some aspects of the immune response, so far there is little indication that hosts with pre-existing immunity are more susceptible to viral infections compared to naïve hosts. However, the tendency of the immune response to focus on previously recognized conserved epitopes when encountering new viral strains can create an opportunity cost when mutations arise in these conserved epitopes. Hosts with different exposure histories may continue to experience distinct patterns of infection over time, which may influence influenza viruses' continued antigenic evolution. Understanding the dynamics of B cell competition that underlie the development of antibody responses might help explain the low effectiveness of current influenza vaccines and lead to better vaccination strategies.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology & Evolution, The University of Chicago, Chicago, IL 19104, USA.
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Saito N, Komori K, Suzuki M, Morimoto K, Kishikawa T, Yasaka T, Ariyoshi K. Negative impact of prior influenza vaccination on current influenza vaccination among people infected and not infected in prior season: A test-negative case-control study in Japan. Vaccine 2016; 35:687-693. [PMID: 28043738 DOI: 10.1016/j.vaccine.2016.11.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/07/2016] [Accepted: 11/07/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND Accumulating evidences indicate that repeated influenza vaccination has negative impact on the vaccine effectiveness (VE). However no published studies considered past influenza infection when assessing the VE of repeated vaccination. METHODS Prospective surveillance was conducted from 2009 to 2012 at a community hospital on a small island in Japan. The study included all outpatients with an influenza-like illness (ILI) who attended the hospital, and a rapid diagnostic test (RDT) was used to diagnose influenza A/B infection. The VE of trivalent inactivated influenza vaccine (TIV) against medically attended influenza A (MA-fluA) was estimated using a test-negative case-control study design. The influence of TIV in the prior season on VE in the current season was investigated in the context of MA-fluA during the prior season. RESULTS During the three influenza seasons, 5838 ILI episodes (4127 subjects) were analysed. Subjects who had an episode of MA-fluA in the prior season were at a significantly lower risk of MA-fluA in the current season (adjusted odds ratio: 0.38, 95% CI: 0.30-0.50). The overall adjusted VE was 28% (95% CI, 14-40). VE was substantially lower in subjects vaccinated in the prior season compared to those who had not been vaccinated in prior season (19%; 95% CI: 0-35 vs 46%; 95% CI: 26-60, test for interaction, P value <0.05). In subjects who did not have MA-fluA in the prior season showed the attenuation of VE due to repeated vaccination (13%; 95% CI: -7 to 30 vs 44%; 95% CI: 24-59, test for interaction, P<0.05). However this effect was not detected in subjects who had contracted MA-fluA in the prior season. CONCLUSIONS Negative effects of repeated vaccination were significant among those without history of MA-fluA in the prior season.
Collapse
Affiliation(s)
- Nobuo Saito
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki, Japan; Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | | | - Motoi Suzuki
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki, Japan
| | - Kounosuke Morimoto
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki, Japan
| | | | | | - Koya Ariyoshi
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki, Japan; Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
28
|
Kim JH, Reber AJ, Kumar A, Ramos P, Sica G, Music N, Guo Z, Mishina M, Stevens J, York IA, Jacob J, Sambhara S. Non-neutralizing antibodies induced by seasonal influenza vaccine prevent, not exacerbate A(H1N1)pdm09 disease. Sci Rep 2016; 6:37341. [PMID: 27849030 PMCID: PMC5110975 DOI: 10.1038/srep37341] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/25/2016] [Indexed: 11/12/2022] Open
Abstract
The association of seasonal trivalent influenza vaccine (TIV) with increased infection by 2009 pandemic H1N1 (A(H1N1)pdm09) virus, initially observed in Canada, has elicited numerous investigations on the possibility of vaccine-associated enhanced disease, but the potential mechanisms remain largely unresolved. Here, we investigated if prior immunization with TIV enhanced disease upon A(H1N1)pdm09 infection in mice. We found that A(H1N1)pdm09 infection in TIV-immunized mice did not enhance the disease, as measured by morbidity and mortality. Instead, TIV-immunized mice cleared A(H1N1)pdm09 virus and recovered at an accelerated rate compared to control mice. Prior TIV immunization was associated with potent inflammatory mediators and virus-specific CD8 T cell activation, but efficient immune regulation, partially mediated by IL-10R-signaling, prevented enhanced disease. Furthermore, in contrast to suggested pathological roles, pre-existing non-neutralizing antibodies (NNAbs) were not associated with enhanced virus replication, but rather with promoted antigen presentation through FcR-bearing cells that led to potent activation of virus-specific CD8 T cells. These findings provide new insights into interactions between pre-existing immunity and pandemic viruses.
Collapse
Affiliation(s)
- Jin Hyang Kim
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Adrian J Reber
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Amrita Kumar
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Patricia Ramos
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Gabriel Sica
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, 1364 Clifton Rd, N.E. Atlanta, GA 30322, USA
| | - Nedzad Music
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Zhu Guo
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Margarita Mishina
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA.,Batelle Memorial Institute, Atlanta, GA 30322, USA
| | - James Stevens
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Ian A York
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| | - Joshy Jacob
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Center, Emory University, 954 Gatewood Rd, Atlanta, GA, USA
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd, Atlanta, GA 30329, USA
| |
Collapse
|
29
|
Kim JH, Mishina M, Chung JR, Cole KS, Nowalk MP, Martin JM, Spencer S, Flannery B, Zimmerman RK, Sambhara S. Cell-Mediated Immunity Against Antigenically Drifted Influenza A(H3N2) Viruses in Children During a Vaccine Mismatch Season. J Infect Dis 2016; 214:1030-8. [PMID: 27534687 DOI: 10.1093/infdis/jiw311] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/18/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Emergence of antigenically drifted influenza A(H3N2) viruses resulted in reduced vaccine effectiveness in all age groups during the 2014-2015 influenza season. In children, inactivated influenza vaccine (IIV) elicited neutralizing antibodies (Abs) against drifted strains at significantly lower levels than against the vaccine strain. Little is known about the cross-reactivity of cell-mediated immunity against drifted strains in children. METHODS Children aged 3-17 years (n = 48) received IIV during the 2014-2015 influenza season. Peripheral blood mononuclear cells, collected before (on day 0) and after (on days 7 and 21) vaccination were evaluated for induction of cross-reactive plasmablasts, memory B cells, and cytokine-secreting CD4(+) and CD8(+) T cells against the vaccine and drifted A(H3N2) viruses by an enzyme-linked immunospot assay and flow cytometry. RESULTS IIV increased frequencies of plasmablasts and memory B cells. The overall induction of the T-cell response was not significant. Both B-cell and T-cell responses showed significant cross-reactivity against A(H3N2) viruses. Age and preexisting immunity affected virus-specific plasmablast responses and fold-change of T-cell responses, respectively. The proportion of T-helper type 1-prone (ie, interferon γ- or tumor necrosis factor α-secreting) CD4(+) T cell responses also increased with age. CONCLUSIONS In children aged 3-17 years, B- and T-cell responses following IIV receipt showed significant cross-reactivity against A(H3N2) viruses during a vaccine mismatch season.
Collapse
Affiliation(s)
- Jin Hyang Kim
- Influenza Division, Centers for Disease Control and Prevention
| | | | | | | | | | - Judith M Martin
- Department of Pediatrics, University of Pittsburgh Children's Hospital of Pittsburgh, Pennsylvania
| | | | | | | | | |
Collapse
|