1
|
Schewe KE, Cooper S, Crowe J, Llewellyn S, Ritter L, Ryan KA, Dibben O. An Optimised Live Attenuated Influenza Vaccine Ferret Efficacy Model Successfully Translates H1N1 Clinical Data. Vaccines (Basel) 2024; 12:1275. [PMID: 39591178 PMCID: PMC11598904 DOI: 10.3390/vaccines12111275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Between 2013 and 2016, the A/H1N1pdm09 component of the live attenuated influenza vaccine (LAIV) produced instances of lower-than-expected vaccine effectiveness. Standard pre-clinical ferret models, using a human-like vaccine dose and focusing on antigenic match to circulating wildtype (wt) strains, were unable to predict these fluctuations. By optimising the vaccine dose and utilising clinically relevant endpoints, we aimed to develop a ferret efficacy model able to reproduce clinical observations. Ferrets were intranasally vaccinated with 4 Log10 FFU/animal (1000-fold reduction compared to clinical dose) of seven historical LAIV formulations with known (19-90%) H1N1 vaccine efficacy or effectiveness (VE). Following homologous H1N1 wt virus challenge, protection was assessed based on primary endpoints of wt virus shedding in the upper respiratory tract and the development of fever. LAIV formulations with high (82-90%) H1N1 VE provided significant protection from wt challenge, while formulations with reduced (19-32%) VE tended not to provide significant protection. The strongest correlation observed was between reduction in wt shedding and VE (R2 = 0.75). Conversely, serum immunogenicity following vaccination was not a reliable indicator of protection (R2 = 0.37). This demonstrated that, by optimisation of the vaccine dose and the use of non-serological, clinically relevant protection endpoints, the ferret model could successfully translate clinical H1N1 LAIV VE data.
Collapse
Affiliation(s)
- Katarzyna E. Schewe
- Flu-BPD, BioPharmaceutical Development, R&D, AstraZeneca, Liverpool L24 9JW, UK; (K.E.S.); (S.C.); (J.C.); (S.L.); (L.R.)
| | - Shaun Cooper
- Flu-BPD, BioPharmaceutical Development, R&D, AstraZeneca, Liverpool L24 9JW, UK; (K.E.S.); (S.C.); (J.C.); (S.L.); (L.R.)
| | - Jonathan Crowe
- Flu-BPD, BioPharmaceutical Development, R&D, AstraZeneca, Liverpool L24 9JW, UK; (K.E.S.); (S.C.); (J.C.); (S.L.); (L.R.)
| | - Steffan Llewellyn
- Flu-BPD, BioPharmaceutical Development, R&D, AstraZeneca, Liverpool L24 9JW, UK; (K.E.S.); (S.C.); (J.C.); (S.L.); (L.R.)
| | - Lydia Ritter
- Flu-BPD, BioPharmaceutical Development, R&D, AstraZeneca, Liverpool L24 9JW, UK; (K.E.S.); (S.C.); (J.C.); (S.L.); (L.R.)
| | - Kathryn A. Ryan
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK;
| | - Oliver Dibben
- Flu-BPD, BioPharmaceutical Development, R&D, AstraZeneca, Liverpool L24 9JW, UK; (K.E.S.); (S.C.); (J.C.); (S.L.); (L.R.)
| |
Collapse
|
2
|
Bissett C, Belij-Rammerstorfer S, Ulaszewska M, Smith H, Kailath R, Morris S, Powers C, Sebastian S, Sharpe HR, Allen ER, Wang Z, Cunliffe RF, Sallah HJ, Spencer AJ, Gilbert S, Tregoning JS, Lambe T. Systemic prime mucosal boost significantly increases protective efficacy of bivalent RSV influenza viral vectored vaccine. NPJ Vaccines 2024; 9:118. [PMID: 38926455 PMCID: PMC11208422 DOI: 10.1038/s41541-024-00912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Although licensed vaccines against influenza virus have been successful in reducing pathogen-mediated disease, they have been less effective at preventing viral infection of the airways and current seasonal updates to influenza vaccines do not always successfully accommodate viral drift. Most licensed influenza and recently licensed RSV vaccines are administered via the intramuscular route. Alternative immunisation strategies, such as intranasal vaccinations, and "prime-pull" regimens, may deliver a more sterilising form of protection against respiratory viruses. A bivalent ChAdOx1-based vaccine (ChAdOx1-NP + M1-RSVF) encoding conserved nucleoprotein and matrix 1 proteins from influenza A virus and a modified pre-fusion stabilised RSV A F protein, was designed, developed and tested in preclinical animal models. The aim was to induce broad, cross-protective tissue-resident T cells against heterotypic influenza viruses and neutralising antibodies against RSV in the respiratory mucosa and systemically. When administered via an intramuscular prime-intranasal boost (IM-IN) regimen in mice, superior protection was generated against challenge with either RSV A, Influenza A H3N2 or H1N1. These results support further clinical development of a pan influenza & RSV vaccine administered in a prime-pull regimen.
Collapse
Affiliation(s)
- Cameron Bissett
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | | | - Marta Ulaszewska
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Holly Smith
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Reshma Kailath
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Claire Powers
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Sebastian
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah R Sharpe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Elizabeth R Allen
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, London, UK
| | - Robert F Cunliffe
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Alexandra J Spencer
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Sarah Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Wang D, Deng Y, Zhou J, Wang W, Huang B, Wang W, Wei L, Ren J, Han R, Bing J, Zhai C, Guo X, Tan W. Single-Dose Intranasal Immunisation with Novel Chimeric H1N1 Expressing the Receptor-Binding Domain of SARS-CoV-2 Induces Robust Mucosal Immunity, Tissue-Resident Memory T Cells, and Heterologous Protection in Mice. Vaccines (Basel) 2023; 11:1453. [PMID: 37766130 PMCID: PMC10537001 DOI: 10.3390/vaccines11091453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Current COVID-19 vaccines can effectively reduce disease severity and hospitalisation; however, they are not considerably effective in preventing infection and transmission. In this context, mucosal vaccines are pertinent to prevent SARS-CoV-2 infection and spread. In this study, we generated a replication-competent recombinant chimeric influenza A virus (IAV) expressing the receptor-binding domain (RBD) of a SARS-CoV-2 prototype in the C-terminus of the neuraminidase (NA) of A/Puerto Rico/08/1934 H1N1 (PR8). The remaining seven segments from A/WSN/1933 H1N1 (WSN) were named PR8NARBD/WSN. We observed that the recombinant virus with the WSN backbone demonstrated improved expression of NA and RBD. A single intranasal dose of PR8NARBD/WSN(103PFU) in mice generated robust mucosal immunity, neutralising antibodies, cellular immunity, and tissue-resident memory T cells specific to SARS-CoV-2 and IAV. Importantly, immunisation with PR8NARBD/WSN viruses effectively protected mice against lethal challenges with H1N1, H3N2 IAV, and SARS-CoV-2 Beta variant and significantly reduced lung viral loads. Overall, our research demonstrates the promising potential of PR8NARBD/WSN as an attractive vaccine against emerging SARS-CoV-2 variants and influenza A virus infections.
Collapse
Affiliation(s)
- Donghong Wang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Yao Deng
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Jianfang Zhou
- State Key Laboratory for Molecular Virology and Genetic Engineering, Chinese National Influenza Center, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China
| | - Wen Wang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Baoying Huang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Wenling Wang
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Lan Wei
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Jiao Ren
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Ruiwen Han
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jialuo Bing
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| | - Chengcheng Zhai
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Xiaoyan Guo
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
| | - Wenjie Tan
- Key Laboratory of Biosafety, National Health Commissions, National Institute for Viral Disease Control and Prevention, China CDC, 155 Changbai Road, Beijing 102206, China; (D.W.)
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
4
|
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
|
5
|
Bull MB, Cohen CA, Leung NH, Valkenburg SA. Universally Immune: How Infection Permissive Next Generation Influenza Vaccines May Affect Population Immunity and Viral Spread. Viruses 2021; 13:1779. [PMID: 34578360 PMCID: PMC8472936 DOI: 10.3390/v13091779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022] Open
Abstract
Next generation influenza vaccines that target conserved epitopes are becoming a clinical reality but still have challenges to overcome. Universal next generation vaccines are considered a vital tool to combat future pandemic viruses and have the potential to vastly improve long-term protection against seasonal influenza viruses. Key vaccine strategies include HA-stem and T cell activating vaccines; however, they could have unintended effects for virus adaptation as they recognise the virus after cell entry and do not directly block infection. This may lead to immune pressure on residual viruses. The potential for immune escape is already evident, for both the HA stem and T cell epitopes, and mosaic approaches for pre-emptive immune priming may be needed to circumvent key variants. Live attenuated influenza vaccines have not been immunogenic enough to boost T cells in adults with established prior immunity. Therefore, viral vectors or peptide approaches are key to harnessing T cell responses. A plethora of viral vector vaccines and routes of administration may be needed for next generation vaccine strategies that require repeated long-term administration to overcome vector immunity and increase our arsenal against diverse influenza viruses.
Collapse
Affiliation(s)
- Maireid B. Bull
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, China; (M.B.B.); (C.A.C.)
| | - Carolyn A. Cohen
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, China; (M.B.B.); (C.A.C.)
| | - Nancy H.L. Leung
- World Health Organization Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, The University of Hong Kong, Hong Kong, China;
| | - Sophie A. Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, China; (M.B.B.); (C.A.C.)
| |
Collapse
|
6
|
Jang YH, Seong BL. Immune Responses Elicited by Live Attenuated Influenza Vaccines as Correlates of Universal Protection against Influenza Viruses. Vaccines (Basel) 2021; 9:vaccines9040353. [PMID: 33916924 PMCID: PMC8067561 DOI: 10.3390/vaccines9040353] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023] Open
Abstract
Influenza virus infection remains a major public health challenge, causing significant morbidity and mortality by annual epidemics and intermittent pandemics. Although current seasonal influenza vaccines provide efficient protection, antigenic changes of the viruses often significantly compromise the protection efficacy of vaccines, rendering most populations vulnerable to the viral infection. Considerable efforts have been made to develop a universal influenza vaccine (UIV) able to confer long-lasting and broad protection. Recent studies have characterized multiple immune correlates required for providing broad protection against influenza viruses, including neutralizing antibodies, non-neutralizing antibodies, antibody effector functions, T cell responses, and mucosal immunity. To induce broadly protective immune responses by vaccination, various strategies using live attenuated influenza vaccines (LAIVs) and novel vaccine platforms are under investigation. Despite superior cross-protection ability, very little attention has been paid to LAIVs for the development of UIV. This review focuses on immune responses induced by LAIVs, with special emphasis placed on the breadth and the potency of individual immune correlates. The promising prospect of LAIVs to serve as an attractive and reliable vaccine platforms for a UIV is also discussed. Several important issues that should be addressed with respect to the use of LAIVs as UIV are also reviewed.
Collapse
Affiliation(s)
- Yo Han Jang
- Department of Biological Sciences and Biotechnology Major in Bio-Vaccine Engineering, Andong National University, Andong 1375, Korea;
- Vaccine Industry Research Institute, Andong National University, Andong 1375, Korea
| | - Baik L. Seong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
- Vaccine Innovation Technology Alliance (VITAL)-Korea, Yonsei University, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-2-2123-7416
| |
Collapse
|
7
|
Marriott AC, Gooch KE, Brown PJ, Ryan KA, Jones NJ, Merredew N, Wiblin N, Dibben O, Bright H, Hallis B, Whittaker CJ, Carroll MW. Severity of heterosubtypic influenza virus infection in ferrets is reduced by live attenuated influenza vaccine. NPJ Vaccines 2021; 6:43. [PMID: 33782409 PMCID: PMC8007727 DOI: 10.1038/s41541-021-00306-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/02/2021] [Indexed: 11/17/2022] Open
Abstract
Live attenuated influenza vaccine (LAIV) is widely used to protect humans from seasonal influenza infection, particularly in children. In contrast to inactivated vaccines, the LAIV can induce both mucosal and cellular immune responses. Here we show that a single dose of monovalent H1N1pdm09-specific LAIV in the ferret model is fully protective against a subsequent wild-type H1N1pdm09 challenge, and furthermore reduces the severity of disease following challenge with a different influenza A subtype (H3N2). The reduced severity comprised reductions in weight loss and fever, as well as more rapid clearance of virus, compared to non-vaccinated H3N2-challenged ferrets. No H3N2-neutralizing antibodies were detected in vaccinated ferret sera. Rather, heterosubtypic protection correlated with interferon-gamma+ (IFN-γ+) T-cell responses measured in peripheral blood and in lung lymphocytes. The IFN-γ+ cells were cross-reactive to H3N2 virus even when obtained from vaccinated animals that had never been exposed to H3N2 virus. We believe this study provides compelling evidence that the LAIV can provide a significant reduction in infection and symptoms when challenged with heterosubtypic influenza strains not included in the LAIV, highlighting the importance of cross-reactive T-cells in the design of a universal influenza vaccine.
Collapse
Affiliation(s)
- Anthony C Marriott
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK.
| | - Karen E Gooch
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Phillip J Brown
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Kathryn A Ryan
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Nicola J Jones
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Natasha Merredew
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Nathan Wiblin
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | - Oliver Dibben
- Flu-MSAT, Biopharmaceutical Development, R&D, AstraZeneca, Liverpool, UK
| | - Helen Bright
- Flu-MSAT, Biopharmaceutical Development, R&D, AstraZeneca, Liverpool, UK
| | - Bassam Hallis
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| | | | - Miles W Carroll
- National Infection Service, Public Health England, Porton Down, Wiltshire, UK
| |
Collapse
|
8
|
Broadly Reactive H2 Hemagglutinin Vaccines Elicit Cross-Reactive Antibodies in Ferrets Preimmune to Seasonal Influenza A Viruses. mSphere 2021; 6:6/2/e00052-21. [PMID: 33692193 PMCID: PMC8546680 DOI: 10.1128/msphere.00052-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Influenza vaccines have traditionally been tested in naive mice and ferrets. However, humans are first exposed to influenza viruses within the first few years of their lives. Therefore, there is a pressing need to test influenza virus vaccines in animal models that have been previously exposed to influenza viruses before being vaccinated. In this study, previously described H2 computationally optimized broadly reactive antigen (COBRA) hemagglutinin (HA) vaccines (Z1 and Z5) were tested in influenza virus “preimmune” ferret models. Ferrets were infected with historical, seasonal influenza viruses to establish preimmunity. These preimmune ferrets were then vaccinated with either COBRA H2 HA recombinant proteins or wild-type H2 HA recombinant proteins in a prime-boost regimen. A set of naive preimmune or nonpreimmune ferrets were also vaccinated to control for the effects of the multiple different preimmunities. All of the ferrets were then challenged with a swine H2N3 influenza virus. Ferrets with preexisting immune responses influenced recombinant H2 HA-elicited antibodies following vaccination, as measured by hemagglutination inhibition (HAI) and classical neutralization assays. Having both H3N2 and H1N1 immunological memory regardless of the order of exposure significantly decreased viral nasal wash titers and completely protected all ferrets from both morbidity and mortality, including the mock-vaccinated ferrets in the group. While the vast majority of the preimmune ferrets were protected from both morbidity and mortality across all of the different preimmunities, the Z1 COBRA HA-vaccinated ferrets had significantly higher antibody titers and recognized the highest number of H2 influenza viruses in a classical neutralization assay compared to the other H2 HA vaccines. IMPORTANCE H1N1 and H3N2 influenza viruses have cocirculated in the human population since 1977. Nearly every human alive today has antibodies and memory B and T cells against these two subtypes of influenza viruses. H2N2 influenza viruses caused the 1957 global pandemic and people born after 1968 have never been exposed to H2 influenza viruses. It is quite likely that a future H2 influenza virus could transmit within the human population and start a new global pandemic, since the majority of people alive today are immunologically naive to viruses of this subtype. Therefore, an effective vaccine for H2 influenza viruses should be tested in an animal model with previous exposure to influenza viruses that have circulated in humans. Ferrets were infected with historical influenza A viruses to more accurately mimic the immune responses in people who have preexisting immune responses to seasonal influenza viruses. In this study, preimmune ferrets were vaccinated with wild-type (WT) and COBRA H2 recombinant HA proteins in order to examine the effects that preexisting immunity to seasonal human influenza viruses have on the elicitation of broadly cross-reactive antibodies from heterologous vaccination.
Collapse
|
9
|
Puksuriwong S, Ahmed MS, Sharma R, Krishnan M, Leong S, Lambe T, McNamara PS, Gilbert SC, Zhang Q. Modified Vaccinia Ankara-Vectored Vaccine Expressing Nucleoprotein and Matrix Protein 1 (M1) Activates Mucosal M1-Specific T-Cell Immunity and Tissue-Resident Memory T Cells in Human Nasopharynx-Associated Lymphoid Tissue. J Infect Dis 2021; 222:807-819. [PMID: 31740938 PMCID: PMC7399703 DOI: 10.1093/infdis/jiz593] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
Background Increasing evidence supports a critical role of CD8+ T-cell immunity against influenza. Activation of mucosal CD8+ T cells, particularly tissue-resident memory T (TRM) cells recognizing conserved epitopes would mediate rapid and broad protection. Matrix protein 1 (M1) is a well-conserved internal protein. Methods We studied the capacity of modified vaccinia Ankara (MVA)–vectored vaccine expressing nucleoprotein (NP) and M1 (MVA-NP+M1) to activate M1-specific CD8+ T-cell response, including TRM cells, in nasopharynx-associated lymphoid tissue from children and adults. Results After MVA-NP+M1 stimulation, M1 was abundantly expressed in adenotonsillar epithelial cells and B cells. MVA-NP+M1 activated a marked interferon γ–secreting T-cell response to M1 peptides. Using tetramer staining, we showed the vaccine activated a marked increase in M158–66 peptide-specific CD8+ T cells in tonsillar mononuclear cells of HLA-matched individuals. We also demonstrated MVA-NP+M1 activated a substantial increase in TRM cells exhibiting effector memory T-cell phenotype. On recall antigen recognition, M1-specific T cells rapidly undergo cytotoxic degranulation, release granzyme B and proinflammatory cytokines, leading to target cell killing. Conclusions MVA-NP+M1 elicits a substantial M1-specific T-cell response, including TRM cells, in nasopharynx-associated lymphoid tissue, demonstrating its strong capacity to expand memory T-cell pool exhibiting effector memory T-cell phenotype, therefore offering great potential for rapid and broad protection against influenza reinfection.
Collapse
Affiliation(s)
- Suttida Puksuriwong
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Muhammad S Ahmed
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Ravi Sharma
- ENT Departments, Alder Hey Children's Hospital, Liverpool, United Kingdom
| | - Madhan Krishnan
- ENT Departments, Alder Hey Children's Hospital, Liverpool, United Kingdom
| | - Sam Leong
- ENT Departments, Aintree University Hospital, Liverpool, United Kingdom
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Paul S McNamara
- Institute of Child Health, Alder Hey Children's Hospital, Liverpool, United Kingdom
| | - Sarah C Gilbert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Qibo Zhang
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
10
|
Mohn KGI, Brokstad KA, Islam S, Oftung F, Tøndel C, Aarstad HJ, Cox RJ. Early Induction of Cross-Reactive CD8+ T-Cell Responses in Tonsils After Live-Attenuated Influenza Vaccination in Children. J Infect Dis 2021; 221:1528-1537. [PMID: 32255493 PMCID: PMC7137893 DOI: 10.1093/infdis/jiz583] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/10/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Live-attenuated influenza vaccine (LAIV) was licensed for prophylaxis of children 2-17 years old in Europe in 2012 and is administered as a nasal spray. Live-attenuated influenza vaccine induces both mucosal and systemic antibodies and systemic T-cell responses. Tonsils are the lymph nodes serving the upper respiratory tract, acting as both induction and effector site for mucosal immunity. METHODS Here, we have studied the early tonsillar T-cell responses induced in children after LAIV. Thirty-nine children were immunized with trivalent LAIV (containing A/H1N1, A/H3N2, and B viruses) at days 3, 7, and 14 before tonsillectomy. Nonvaccinated controls were included for comparison. Tonsils and peripheral blood (pre- and postvaccination) were collected to study T-cell responses. RESULTS Tonsillar and systemic T-cell responses differed between influenza strains, and both were found against H3N2 and B viruses, whereas only systemic responses were observed against A/H1N1. A significant increase in cross-reactive tonsillar CD8+ T cells recognizing conserved epitopes from a broad range of seasonal and pandemic viruses occurred at day 14. Tonsillar T cells showed significant cytokine responses (Th1, Th2, and granulocyte-macrophage colony-stimulating factor). CONCLUSIONS Our findings support the use of LAIV in children to elicit broadly cross-reactive T cells, which are not induced by traditional inactivated influenza vaccines and may provide protection to novel virus strains.
Collapse
Affiliation(s)
- K G-I Mohn
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| | - K A Brokstad
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - S Islam
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway
| | - F Oftung
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - C Tøndel
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - H J Aarstad
- Department of Otorhinolaryngology/Head and Neck Surgery, Haukeland University Hospital, Bergen, Norway
| | - R J Cox
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
11
|
Martini V, Paudyal B, Chrun T, McNee A, Edmans M, Atangana Maze E, Clark B, Nunez A, Dolton G, Sewell A, Beverley P, MacLoughlin R, Townsend A, Tchilian E. Simultaneous Aerosol and Intramuscular Immunization with Influenza Vaccine Induces Powerful Protective Local T Cell and Systemic Antibody Immune Responses in Pigs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:652-663. [PMID: 33328212 PMCID: PMC7812058 DOI: 10.4049/jimmunol.2001086] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 01/04/2023]
Abstract
A vaccine providing both powerful Ab and cross-reactive T cell immune responses against influenza viruses would be beneficial for both humans and pigs. In this study, we evaluated i.m., aerosol (Aer), and simultaneous systemic and respiratory immunization (SIM) by both routes in Babraham pigs, using the single cycle candidate influenza vaccine S-FLU. After prime and boost immunization, pigs were challenged with H1N1pdm09 virus. i.m.-immunized pigs generated a high titer of neutralizing Abs but poor T cell responses, whereas Aer induced powerful respiratory tract T cell responses but a low titer of Abs. SIM pigs combined high Ab titers and strong local T cell responses. SIM showed the most complete suppression of virus shedding and the greatest improvement in pathology. We conclude that SIM regimes for immunization against respiratory pathogens warrant further study.
Collapse
Affiliation(s)
- Veronica Martini
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom; .,Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Basu Paudyal
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Tiphany Chrun
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Adam McNee
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Matthew Edmans
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | | | - Beckie Clark
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom
| | - Alejandro Nunez
- UK Animal and Plant Health Agency-Weybridge, New Haw, Addlestone KT15 3NB, United Kingdom
| | - Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Andrew Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Peter Beverley
- National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom; and
| | | | - Alain Townsend
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Elma Tchilian
- The Pirbright Institute, Pirbright GU24 0NF, United Kingdom;
| |
Collapse
|
12
|
Kerstetter LJ, Buckley S, Bliss CM, Coughlan L. Adenoviral Vectors as Vaccines for Emerging Avian Influenza Viruses. Front Immunol 2021; 11:607333. [PMID: 33633727 PMCID: PMC7901974 DOI: 10.3389/fimmu.2020.607333] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
It is evident that the emergence of infectious diseases, which have the potential for spillover from animal reservoirs, pose an ongoing threat to global health. Zoonotic transmission events have increased in frequency in recent decades due to changes in human behavior, including increased international travel, the wildlife trade, deforestation, and the intensification of farming practices to meet demand for meat consumption. Influenza A viruses (IAV) possess a number of features which make them a pandemic threat and a major concern for human health. Their segmented genome and error-prone process of replication can lead to the emergence of novel reassortant viruses, for which the human population are immunologically naïve. In addition, the ability for IAVs to infect aquatic birds and domestic animals, as well as humans, increases the likelihood for reassortment and the subsequent emergence of novel viruses. Sporadic spillover events in the past few decades have resulted in human infections with highly pathogenic avian influenza (HPAI) viruses, with high mortality. The application of conventional vaccine platforms used for the prevention of seasonal influenza viruses, such as inactivated influenza vaccines (IIVs) or live-attenuated influenza vaccines (LAIVs), in the development of vaccines for HPAI viruses is fraught with challenges. These issues are associated with manufacturing under enhanced biosafety containment, and difficulties in propagating HPAI viruses in embryonated eggs, due to their propensity for lethality in eggs. Overcoming manufacturing hurdles through the use of safer backbones, such as low pathogenicity avian influenza viruses (LPAI), can also be a challenge if incompatible with master strain viruses. Non-replicating adenoviral (Ad) vectors offer a number of advantages for the development of vaccines against HPAI viruses. Their genome is stable and permits the insertion of HPAI virus antigens (Ag), which are expressed in vivo following vaccination. Therefore, their manufacture does not require enhanced biosafety facilities or procedures and is egg-independent. Importantly, Ad vaccines have an exemplary safety and immunogenicity profile in numerous human clinical trials, and can be thermostabilized for stockpiling and pandemic preparedness. This review will discuss the status of Ad-based vaccines designed to protect against avian influenza viruses with pandemic potential.
Collapse
Affiliation(s)
- Lucas J. Kerstetter
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Stephen Buckley
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carly M. Bliss
- Division of Cancer & Genetics, Division of Infection & Immunity, School of Medicine, Cardiff University, Wales, United Kingdom
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
13
|
Stepanova E, Krutikova E, Wong PF, Matyushenko V, Bazhenova E, Isakova-Sivak I, Rudenko L. Safety, Immunogenicity, and Protective Efficacy of a Chimeric A/B Live Attenuated Influenza Vaccine in a Mouse Model. Microorganisms 2021; 9:microorganisms9020259. [PMID: 33513862 PMCID: PMC7910998 DOI: 10.3390/microorganisms9020259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/06/2021] [Accepted: 01/25/2021] [Indexed: 01/20/2023] Open
Abstract
Influenza A and B viruses cause significant morbidity and mortality worldwide. Current influenza vaccines are composed of three or four strains: A/H1N1, A/H3N2, and B (Victoria and Yamagata lineages). It is of great interest if immunization against both type A and B influenza viruses can be combined in a single vaccine strain, thus reducing the cost of vaccine production and the possibility of strain interference within the multicomponent vaccine. In the current study, we developed an experimental live cold-adapted influenza intertype reassortant (influenza A and B) vaccine on the live attenuated influenza vaccine (LAIV) A/Leningrad/134/17/57 backbone. Hemagglutinin (HA) and neuraminidase (NA) functional domains were inherited from the influenza B/Brisbane/60/2008 strain, whereas their packaging signals were substituted with appropriate fragments of influenza A virus genes. The recombinant A/B virus efficiently replicated in eggs and Madin–Darby Canine Kidney (MDCK) cells under optimal conditions, temperature-sensitive phenotype was maintained, and its antigenic properties matched the influenza B parental virus. The chimeric vaccine was attenuated in mice: after intranasal immunization, viral replication was seen only in nasal turbinates but not in the lungs. Immunological studies demonstrated the induction of IgG antibody responses against the influenza A and B virus, whereas hemagglutination inhibition (HAI) and neutralizing antibodies were detected only against the influenza B virus, resulting in significant protection of immunized animals against influenza B virus challenge. IFNγ-secreting CD8 effector memory T cells (CD44+CD62L−) were detected in mouse splenocytes after stimulation with the specific influenza A peptide (NP366); however, the T-cell response was not sufficient to protect animals against infection with a high-dose mouse-adapted A/California/07/2009 (H1N1pdm09) virus, most probably due to the mismatch of key T-cell epitopes of the H1N1 virus and the LAIV backbone. Overall, generation of the chimeric A/B LAIV virus on a licensed LAIV backbone demonstrated prospects for the development of safe and efficacious vaccine candidates that afford combined protection against both type A and type B influenza viruses; however, further optimization of the T-cell epitope content within the LAIV backbone may be required.
Collapse
|
14
|
Beijnen EMS, van Haren SD. Vaccine-Induced CD8 + T Cell Responses in Children: A Review of Age-Specific Molecular Determinants Contributing to Antigen Cross-Presentation. Front Immunol 2020; 11:607977. [PMID: 33424857 PMCID: PMC7786054 DOI: 10.3389/fimmu.2020.607977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Infections are most common and most severe at the extremes of age, the young and the elderly. Vaccination can be a key approach to enhance immunogenicity and protection against pathogens in these vulnerable populations, who have a functionally distinct immune system compared to other age groups. More than 50% of the vaccine market is for pediatric use, yet to date vaccine development is often empiric and not tailored to molecular distinctions in innate and adaptive immune activation in early life. With modern vaccine development shifting from whole-cell based vaccines to subunit vaccines also comes the need for formulations that can elicit a CD8+ T cell response when needed, for example, by promoting antigen cross-presentation. While our group and others have identified many cellular and molecular determinants of successful activation of antigen-presenting cells, B cells and CD4+ T cells in early life, much less is known about the ontogeny of CD8+ T cell induction. In this review, we summarize the literature pertaining to the frequency and phenotype of newborn and infant CD8+ T cells, and any evidence of induction of CD8+ T cells by currently licensed pediatric vaccine formulations. In addition, we review the molecular determinants of antigen cross-presentation on MHC I and successful CD8+ T cell induction and discuss potential distinctions that can be made in children. Finally, we discuss recent advances in development of novel adjuvants and provide future directions for basic and translational research in this area.
Collapse
Affiliation(s)
- Elisabeth M. S. Beijnen
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Simon D. van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Roy S, Williams CM, Wijesundara DK, Furuya Y. Impact of Pre-Existing Immunity to Influenza on Live-Attenuated Influenza Vaccine (LAIV) Immunogenicity. Vaccines (Basel) 2020; 8:E683. [PMID: 33207559 PMCID: PMC7711626 DOI: 10.3390/vaccines8040683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022] Open
Abstract
During the previous influenza seasons, between 2010 and 2016, the live attenuated influenza vaccine (LAIV) provided variable efficacy against influenza in the U.S., causing the recommendation against the use of the LAIV. In striking contrast, pre-clinical studies have repeatedly demonstrated superior efficacy of LAIV against mismatched influenza viruses, compared to inactivated influenza vaccines (IIV). This disparity in reported vaccine efficacies between pre-clinical and clinical studies may in part be explained by limitations of the animal models of influenza. In particular, the absence of pre-existing immunity in animal models has recently emerged as a potential explanation for the discrepancies between preclinical findings and human studies. This commentary focuses on the potential impact of pre-existing immunity on LAIV induced immunogenicity with an emphasis on cross-protective immunity.
Collapse
Affiliation(s)
- Sreeja Roy
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| | - Clare M. Williams
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| | - Danushka K. Wijesundara
- The School of Chemistry and Molecular Biosciences, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland 4072, Australia;
| | - Yoichi Furuya
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| |
Collapse
|
16
|
Stambas J, Lu C, Tripp RA. Innate and adaptive immune responses in respiratory virus infection: implications for the clinic. Expert Rev Respir Med 2020; 14:1141-1147. [PMID: 32762572 DOI: 10.1080/17476348.2020.1807945] [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] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The innate immune response is the first line of defense and consists of physical, chemical and cellular defenses. The adaptive immune response is the second line of defense and is pathogen-specific. Innate immunity occurs immediately while adaptive immunity develops upon pathogen exposure, and is long-lasting, highly specific, and sustained by memory T cells. Respiratory virus infection typically induces effective immunity but over-exuberant responses are associated with pathophysiology. Cytokines expressed in response to viral infection can enhance biological responses, activate, and trigger signaling pathways leading to adaptive immunity Vaccines induce immunity, specifically B and T cell responses. Vaccination is generally efficacious, but for many viruses, our understanding of vaccination strategies and immunity is incomplete or in its infancy. Studies that examine innate and adaptive immune responses to respiratory virus infection will aid vaccine development and may reduce the burden of respiratory viral disease. AREAS COVERED A literature search was performed using PubMed. The search covered: innate, adaptive, respiratory virus, vaccine development, B cell, and T cell. EXPERT OPINION Immunity rests on two pillars, i.e. the innate and adaptive immune system, which function together on different tasks to maintain homeostasis. a better understanding of immunity is necessary for disease prevention and intervention.
Collapse
Affiliation(s)
- John Stambas
- School of Medicine, Deakin University , Melbourne, Australia
| | - Chunni Lu
- School of Medicine, Deakin University , Melbourne, Australia
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia , Athens, GA, USA
| |
Collapse
|
17
|
Loes AN, Gentles LE, Greaney AJ, Crawford KHD, Bloom JD. Attenuated Influenza Virions Expressing the SARS-CoV-2 Receptor-Binding Domain Induce Neutralizing Antibodies in Mice. Viruses 2020; 12:E987. [PMID: 32899480 PMCID: PMC7552029 DOI: 10.3390/v12090987] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/20/2022] Open
Abstract
An effective vaccine is essential for controlling the spread of the SARS-CoV-2 virus. Here, we describe an influenza virus-based vaccine for SARS-CoV-2. We incorporated a membrane-anchored form of the SARS-CoV-2 spike receptor binding domain (RBD) in place of the neuraminidase (NA) coding sequence in an influenza virus also possessing a mutation that reduces the affinity of hemagglutinin for its sialic acid receptor. The resulting ΔNA(RBD)-Flu virus can be generated by reverse genetics and grown to high titers in cell culture. A single-dose intranasal inoculation of mice with ΔNA(RBD)-Flu elicits serum neutralizing antibody titers against SAR-CoV-2 comparable to those observed in humans following natural infection (~1:200). Furthermore, ΔNA(RBD)-Flu itself causes no apparent disease in mice. It might be possible to produce a vaccine similar to ΔNA(RBD)-Flu at scale by leveraging existing platforms for the production of influenza vaccines.
Collapse
Affiliation(s)
- Andrea N. Loes
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
| | - Lauren E. Gentles
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
| | - Allison J. Greaney
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Katharine H. D. Crawford
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Jesse D. Bloom
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
18
|
Loes AN, Gentles LE, Greaney AJ, Crawford KHD, Bloom JD. Attenuated influenza virions expressing the SARS-CoV-2 receptor-binding domain induce neutralizing antibodies in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.08.12.248823. [PMID: 32817935 PMCID: PMC7430565 DOI: 10.1101/2020.08.12.248823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
An effective vaccine is essential to controlling the spread of SARS-CoV-2 virus. Here, we describe an influenza-virus-based vaccine for SARS-CoV-2. We incorporated a membrane-anchored form of the SARS-CoV-2 Spike receptor binding domain (RBD) in place of the neuraminidase (NA) coding sequence in an influenza virus also possessing a mutation that reduces the affinity of hemagglutinin for its sialic acid receptor. The resulting ΔNA(RBD)-Flu virus can be generated by reverse genetics and grown to high titers in cell culture. A single-dose intranasal inoculation of mice with ΔNA(RBD)-Flu elicits serum neutralizing antibody titers against SAR-CoV-2 comparable to those observed in humans following natural infection (~1:200). Furthermore, ΔNA(RBD)-Flu itself causes no apparent disease in mice. It might be possible to produce a vaccine similar to ΔNA(RBD)-Flu at scale by leveraging existing platforms for production of influenza vaccines.
Collapse
Affiliation(s)
- Andrea N. Loes
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
| | - Lauren E. Gentles
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
| | - Allison J. Greaney
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Katharine H. D. Crawford
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Jesse D. Bloom
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
19
|
Lartey S, Zhou F, Brokstad KA, Mohn KGI, Slettevoll SA, Pathirana RD, Cox RJ. Live-Attenuated Influenza Vaccine Induces Tonsillar Follicular T Helper Cell Responses That Correlate With Antibody Induction. J Infect Dis 2020; 221:21-32. [PMID: 31250024 PMCID: PMC6910880 DOI: 10.1093/infdis/jiz321] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/25/2019] [Indexed: 11/14/2022] Open
Abstract
Background Influenza remains a major threat to public health. Live-attenuated influenza vaccines (LAIV) have been shown to be effective, particularly in children. Follicular T helper (TFH) cells provide B-cell help and are crucial for generating long-term humoral immunity. However the role of TFH cells in LAIV-induced immune responses is unknown. Methods We collected tonsils, plasma, and saliva samples from children and adults receiving LAIV prior to tonsillectomy. We measured influenza-specific TFH-cell responses after LAIV by flow cytometry and immunohistochemistry. Systemic and local antibody responses were analysed by hemagglutination inhibition assay and enzyme-linked immunosorbent assay. Results We report that LAIV induced early (3–7 days post-vaccination) activation of tonsillar follicles and influenza-specific TFH-cell (CXCR5+CD57+CD4+ T cell) responses in children, and to a lesser extent in adults. Serological analyses showed that LAIV elicited rapid (day 14) and long-term (up to 1 year post-vaccination) antibody responses (hemagglutination inhibition, influenza-specific IgG) in children, but not adults. There was an inverse correlation between pre-existing influenza-specific salivary IgA concentrations and tonsillar TFH-cell responses, and a positive correlation between tonsillar TFH-cell and systemic IgG induction after LAIV. Conclusions Our data, taken together, demonstrate an important role of tonsillar TFH cells in LAIV-induced immunity in humans.
Collapse
Affiliation(s)
| | - Fan Zhou
- Influenza Center, Bergen, Norway.,K.G. Jebsen Center for Influenza Vaccines Research, Bergen, Norway
| | - Karl A Brokstad
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | | | | | - Rebecca J Cox
- Influenza Center, Bergen, Norway.,K.G. Jebsen Center for Influenza Vaccines Research, Bergen, Norway.,Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
20
|
Matyushenko V, Kotomina T, Kudryavtsev I, Mezhenskaya D, Prokopenko P, Matushkina A, Sivak K, Muzhikyan A, Rudenko L, Isakova-Sivak I. Conserved T-cell epitopes of respiratory syncytial virus (RSV) delivered by recombinant live attenuated influenza vaccine viruses efficiently induce RSV-specific lung-localized memory T cells and augment influenza-specific resident memory T-cell responses. Antiviral Res 2020; 182:104864. [PMID: 32585323 PMCID: PMC7313889 DOI: 10.1016/j.antiviral.2020.104864] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/30/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Respiratory syncytial virus (RSV) can cause recurrent infection in people because it does not stimulate a long-lived immunological memory. There is an urgent need to develop a safe and efficacious vaccine against RSV that would induce immunological memory without causing immunopathology following natural RSV infection. We have previously generated two recombinant live attenuated influenza vaccine (LAIV) viruses that encode immunodominant T-cell epitopes of RSV M2 protein in the neuraminidase or NS1 genes. These chimeric vaccines afforded protection against influenza and RSV infection in mice, without causing pulmonary eosinophilia or inflammatory RSV disease. The current study assessed the formation of influenza-specific and RSV-specific CD4 and CD8 T-cell responses in the lungs of mice, with special attention to the lung tissue-resident memory T cell subsets (TRM). The RSV epitopes did not affect influenza-specific CD4 effector memory T cell (Tem) levels in the lungs. The majority of these cells formed by LAIV or LAIV-RSV viruses had CD69+CD103- phenotype. Both LAIV+NA/RSV and LAIV+NS/RSV recombinant viruses induced significant levels of RSV M282 epitope-specific lung-localized CD8 Tem cells expressing both CD69 and CD103 TRM markers. Surprisingly, the CD69+CD103+ influenza-specific CD8 Tem responses were augmented by the addition of RSV epitopes, possibly as a result of the local microenvironment formed by the RSV-specific memory T cells differentiating to TRM in the lungs of mice immunized with LAIV-RSV chimeric viruses. This study provides evidence that LAIV vector-based vaccination can induce robust lung-localized T-cell immunity to the inserted T-cell epitope of a foreign pathogen, without altering the immunogenicity of the viral vector itself. Two LAIV-RSV vaccine viruses induced RSV M282-specific effector memory CD8 T cells producing both IFNγ and TNFα cytokines. The inserted RSV epitopes did not affect influenza-specific CD4 Tem levels in the lungs of immunized mice. LAIV-RSV viruses induced RSV M282-specific lung-localized CD8 Tem cells expressing both CD69 and CD103 TRM markers. The magnitude of RSV M282-specific CD8 Tem responses correlates with protection against RSV-induced lung pathology. The addition of RSV epitopes into the LAIV strain augmented CD69+CD103+ influenza-specific CD8 Tem responses in the lungs.
Collapse
Affiliation(s)
- Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Polina Prokopenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Anastasia Matushkina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Konstantin Sivak
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Arman Muzhikyan
- Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg, Russia.
| |
Collapse
|
21
|
The Effects of Pre-Existing Antibodies on Live-Attenuated Viral Vaccines. Viruses 2020; 12:v12050520. [PMID: 32397218 PMCID: PMC7290594 DOI: 10.3390/v12050520] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Live-attenuated vaccines (LAVs) have achieved remarkable successes in controlling virus spread, as well as for other applications such as cancer immunotherapy. However, with rapid increases in international travel, globalization, geographic spread of viral vectors, and widespread use of vaccines, there is an increasing need to consider how pre-exposure to viruses which share similar antigenic regions can impact vaccine efficacy. Pre-existing antibodies, derived from either from maternal–fetal transmission, or by previous infection or vaccination, have been demonstrated to interfere with vaccine immunogenicity of measles, adenovirus, and influenza LAVs. Immune interference of LAVs can be caused by the formation of virus–antibody complexes that neutralize virus infection in antigen-presenting cells, or by the cross-linking of the B-cell receptor with the inhibitory receptor, FcγRIIB. On the other hand, pre-existing antibodies can augment flaviviral LAV efficacy such as that of dengue and yellow fever virus, especially when pre-existing antibodies are present at sub-neutralizing levels. The increased vaccine immunogenicity can be facilitated by antibody-dependent enhancement of virus infection, enhancing virus uptake in antigen-presenting cells, and robust induction of innate immune responses that promote vaccine immunogenicity. This review examines the literature on this topic and examines the circumstances where pre-existing antibodies can inhibit or enhance LAV efficacy. A better knowledge of the underlying mechanisms involved could allow us to better manage immunization in seropositive individuals and even identify possibilities that could allow us to exploit pre-existing antibodies to boost vaccine-induced responses for improved vaccine efficacy.
Collapse
|
22
|
Isakova-Sivak I, Grigorieva E, Rudenko L. Insights into current clinical research on the immunogenicity of live attenuated influenza vaccines. Expert Rev Vaccines 2020; 19:43-55. [PMID: 31903816 DOI: 10.1080/14760584.2020.1711056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction: Live attenuated influenza vaccines (LAIVs) have been in use for more than three decades and are now licensed in many countries. There is evidence that LAIVs can have greater efficacy than inactivated influenza vaccines, especially against mismatched influenza, however, in recent years, a number of trials have found a lack of LAIV efficacy, mainly in relation to the H1N1 virus.Areas covered: In this review, we summarize the results of clinical research published in the past 5 years on the immunogenicity of LAIVs, with special attention to the mechanisms of establishing protective immunity and some factors that may influence immunogenicity and efficacy.Expert opinion: A number of recent clinical studies confirmed that the immune responses to LAIVs are multifaceted, involving different immune mechanisms. These trials suggest that the intrinsic replicative properties of each LAIV component should be taken into account, and the precise effects of adding a fourth vaccine strain to trivalent LAIV formulations are still to be identified. In addition, new data are emerging regarding the impact of pre-vaccination conditions, such as preexisting immunity or concurrent asymptomatic viral and bacterial respiratory infections, on LAIV immunogenicity, suggesting the importance of monitoring them during clinical trials or vaccination campaigns.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Elena Grigorieva
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
23
|
Abstract
Introduction: High variance is associated with influenza vaccine effectiveness (VE). Accumulating evidence suggests that preexisting influenza-specific immunity results in the variance in VE and skews overall immune response to vaccination. Nevertheless, the investigation of preexisting immunity is highly limited due to the lack of proper methodology to explore the complex individual immune history.Areas covered: Retrospective observational studies have shown that the preexisting influenza specific immunity influences on VE. To simplify a discussion, we summarized important findings from the observational studies based on the transition of the individual immune history: the first exposure to influenza virus, the first vaccination, and repetitive exposure throughout life. We also discussed the prospectus of pre-immunized animal models to investigate the interaction between preexisting immunity and vaccine efficacy.Expert opinion: A better understanding in the underlying mechanisms on preexisting immunity is critical to improve VE and to help develop novel vaccine strategies. Using animals pre-immunized with historical influenza strains is a promising approach to verify the underlying immunologic mechanism of interaction between preexisting immunity and vaccine antigen. Also, pre-immunized animal models will be better able to evaluate the efficacy of novel vaccine strategies than naïve animals.
Collapse
Affiliation(s)
- Hyesun Jang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.,Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| |
Collapse
|
24
|
Jang YH, Seong BL. The Quest for a Truly Universal Influenza Vaccine. Front Cell Infect Microbiol 2019; 9:344. [PMID: 31649895 PMCID: PMC6795694 DOI: 10.3389/fcimb.2019.00344] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/24/2019] [Indexed: 12/17/2022] Open
Abstract
There is an unmet public health need for a universal influenza vaccine (UIV) to provide broad and durable protection from influenza virus infections. The identification of broadly protective antibodies and cross-reactive T cells directed to influenza viral targets present a promising prospect for the development of a UIV. Multiple targets for cross-protection have been identified in the stalk and head of hemagglutinin (HA) to develop a UIV. Recently, neuraminidase (NA) has received significant attention as a critical component for increasing the breadth of protection. The HA stalk-based approaches have shown promising results of broader protection in animal studies, and their feasibility in humans are being evaluated in clinical trials. Mucosal immune responses and cross-reactive T cell immunity across influenza A and B viruses intrinsic to live attenuated influenza vaccine (LAIV) have emerged as essential features to be incorporated into a UIV. Complementing the weakness of the stand-alone approaches, prime-boost vaccination combining HA stalk, and LAIV is under clinical evaluation, with the aim to increase the efficacy and broaden the spectrum of protection. Preexisting immunity in humans established by prior exposure to influenza viruses may affect the hierarchy and magnitude of immune responses elicited by an influenza vaccine, limiting the interpretation of preclinical data based on naive animals, necessitating human challenge studies. A consensus is yet to be achieved on the spectrum of protection, efficacy, target population, and duration of protection to define a “universal” vaccine. This review discusses the recent advancements in the development of UIVs, rationales behind cross-protection and vaccine designs, and challenges faced in obtaining balanced protection potency, a wide spectrum of protection, and safety relevant to UIVs.
Collapse
Affiliation(s)
- Yo Han Jang
- Molecular Medicine Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Baik Lin Seong
- Molecular Medicine Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.,Vaccine Translational Research Center, Yonsei University, Seoul, South Korea
| |
Collapse
|
25
|
Islam S, Zhou F, Lartey S, Mohn KGI, Krammer F, Cox RJ, Brokstad KA. Functional immune response to influenza H1N1 in children and adults after live attenuated influenza virus vaccination. Scand J Immunol 2019; 90:e12801. [PMID: 31269273 DOI: 10.1111/sji.12801] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 02/02/2023]
Abstract
Influenza virus is a major respiratory pathogen, and vaccination is the main method of prophylaxis. In 2012, the trivalent live attenuated influenza vaccine (LAIV) was licensed in Europe for use in children. Vaccine-induced antibodies directed against the main viral surface glycoproteins, haemagglutinin (HA) and neuraminidase (NA) play important roles in limiting virus infection. The objective of this study was to dissect the influenza-specific antibody responses in children and adults, and T cell responses in children induced after LAIV immunization to the A/H1N1 virus. Blood samples were collected pre- and at 28 and 56 days post-vaccination from 20 children and 20 adults. No increase in micro-neutralization (MN) antibodies against A/H1N1 was observed after vaccination. A/H1N1 stalk-specific neutralizing and NA-inhibiting (NI) antibodies were boosted in children after LAIV. Interferon γ-producing T cells increased significantly in children, and antibody-dependent cellular-mediated cytotoxic (ADCC) cell activity increased slightly in children after vaccination, although this change was not significant. The results indicate that the NI assay is more sensitive to qualitative changes in serum antibodies after LAIV. There was a considerable difference in the immune response in children and adults after vaccination, which may be related to priming and previous influenza history. Our findings warrant further studies for evaluating LAIV vaccination immunogenicity.
Collapse
Affiliation(s)
- Shahinul Islam
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway
| | - Fan Zhou
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway
| | - Sarah Lartey
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway
| | - Kristin G I Mohn
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebecca Jane Cox
- Department of Clinical Science, Influenza Centre, University of Bergen, Bergen, Norway.,Department of Clinical Science, K.G. Jebsen Centre for Influenza Vaccine Research, University of Bergen, Bergen, Norway.,Department of Research & Development, Haukeland University Hospital, Bergen, Norway
| | - Karl Albert Brokstad
- Department of Clinical Science, Broegelmann Research Laboratory, University of Bergen, Bergen, Norway
| |
Collapse
|
26
|
Comparative Immunogenicity of the 2014-2015 Northern Hemisphere Trivalent IIV and LAIV against Influenza A Viruses in Children. Vaccines (Basel) 2019; 7:vaccines7030087. [PMID: 31408963 PMCID: PMC6789519 DOI: 10.3390/vaccines7030087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 11/23/2022] Open
Abstract
Both inactivated influenza vaccines (IIV) and live-attenuated influenza vaccines (LAIV) have been recommended for administration to children. Children are a high-risk group for severe influenza, and a major source of transmission. Therefore, prevention of infection by vaccination is particularly important. However, efficacy and immunogenicity of these vaccines are known to vary by season and geographic location. We compared the immunogenicity of the 2014–2015 Northern Hemisphere trivalent IIV and LAIV against influenza A virus in Canadian Hutterite children aged 2 to 17 using hemagglutination inhibition (HAI) assays, and enzyme-linked immunosorbent assays to measure hemagglutinin-specific serum IgA and mucosal IgA. Both vaccine formulations induced significant increases in HAI titers against H1N1 and H3N2 vaccine strains. Serum IgA titers against H3N2 were significantly boosted by both IIV and LAIV, while only IIV induced a significant increase in serum IgA specific to the H1N1 vaccine strain. While HAI titers correlated with protection conferred by IIV, mucosal IgA titers correlated with protection conferred by LAIV (mucosal IgA titers could not be established as a correlate for IIV due to sample size limitations). IIV and LAIV were previously reported to be equally efficacious in this cohort, although the immunogenicity of IIV was generally superior.
Collapse
|
27
|
Kotomina T, Isakova-Sivak I, Matyushenko V, Kim KH, Lee Y, Jung YJ, Kang SM, Rudenko L. Recombinant live attenuated influenza vaccine viruses carrying CD8 T-cell epitopes of respiratory syncytial virus protect mice against both pathogens without inflammatory disease. Antiviral Res 2019; 168:9-17. [PMID: 31075351 PMCID: PMC6620116 DOI: 10.1016/j.antiviral.2019.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/29/2022]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory disease in young children, elderly and immunocompromised adults. There is no licensed vaccine against RSV although development of an effective and safe RSV vaccine has been a high priority for several decades. Among the various vaccine platforms, the viral-vectored RSV vaccines based on licensed cold-adapted live attenuated influenza vaccine (LAIV) might offer an advantage of inducing adequate mucosal CD8 T cell immunity at the infection site of respiratory pathogens. We constructed two recombinant LAIV viruses expressing immunodominant T-cell epitopes of RSV M2-1 protein. The results in this study provide evidence that RSV CD8 T cell epitopes delivered by LAIV viral vector could confer protection against RSV infection without causing pulmonary eosinophilia and inflammatory RSV disease in mice. In addition, these chimeric LAIV-RSV vaccines retained their attenuated phenotype and ability to protect against virulent influenza virus, thus providing a unique approach to fight against two dangerous respiratory viral pathogens using a single vaccine preparation.
Collapse
Affiliation(s)
- Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia.
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| |
Collapse
|
28
|
Navarro-Torné A, Hanrahan F, Kerstiëns B, Aguar P, Matthiessen L. Public Health-Driven Research and Innovation for Next-Generation Influenza Vaccines, European Union. Emerg Infect Dis 2019; 25. [PMID: 30666948 PMCID: PMC6346458 DOI: 10.3201/eid2502.180359] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Influenza virus infections are a major public health threat. Vaccination is available, but unpredictable antigenic changes in circulating strains require annual modification of seasonal influenza vaccines. Vaccine effectiveness has proven limited, particularly in certain groups, such as the elderly. Moreover, preparedness for upcoming pandemics is challenging because we can predict neither the strain that will cause the next pandemic nor the severity of the pandemic. The European Union fosters research and innovation to develop novel vaccines that evoke broadly protective and long-lasting immune responses against both seasonal and pandemic influenza, underpinned by a political commitment to global public health.
Collapse
|
29
|
Ong EZ, Gan ES, de Alwis R, Wijaya L, Ong XM, Zhang M, Wong AW, Cheung YB, Zellweger RM, Ooi EE, Low JG. Genomic signature of early T-cell response is associated with lower antibody titer threshold for sterilizing immunity. Antiviral Res 2019; 166:35-41. [PMID: 30940521 DOI: 10.1016/j.antiviral.2019.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/21/2019] [Accepted: 03/27/2019] [Indexed: 02/02/2023]
Abstract
Vaccination is an effective approach to reduce disease burden. High vaccination coverage blocks pathogen transmission to ensure herd immunity. However, the concept of herd immunity assumes that vaccinated individuals cannot be infected and mediate silent pathogen transmission. While the correlates of vaccine-mediated protection against disease have been examined, the correlates of sterilizing immunity that prevents infection have not been systematically defined. Here, we used full genome expression profiling to explore the molecular correlates of serological response and non-response to measles, mumps and rubella (MMR) vaccination as surrogates of infection and sterilizing immunity, respectively. We observed that the antibody titers needed to sterilize infection with the vaccine strains were higher than current WHO disease protection thresholds. In subjects with baseline antibodies below such sterilizing immunity thresholds, serological non-response to MMR vaccination was associated with gene expression profile indicative of early T-cell activation and signalling. Specifically, genes that regulate T-cell function and response were induced at day 1 post-vaccination in non-responders but not in responders. These findings suggest that rapid T-cell response prevented MMR vaccine infection to limit antigenic presentation and hence serological response. Collectively, our findings suggest an important role for T-cells in engendering sterilizing immunity.
Collapse
Affiliation(s)
- Eugenia Z Ong
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Esther S Gan
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Ruklanthi de Alwis
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Limin Wijaya
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Xin Mei Ong
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Abigail Wl Wong
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Yin Bun Cheung
- Center for Quantitative Medicine, Duke-NUS Medical School, Singapore; Department for International Health, University of Tampere, 33100, Finland
| | - Raphaël M Zellweger
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Eng Eong Ooi
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART), Singapore
| | - Jenny G Low
- Viral Research and Experimental Medicine Centre @ SingHealth-Duke NUS, Singapore; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Department of Infectious Diseases, Singapore General Hospital, Singapore.
| |
Collapse
|
30
|
Epstein SL. Universal Influenza Vaccines: Progress in Achieving Broad Cross-Protection In Vivo. Am J Epidemiol 2018; 187:2603-2614. [PMID: 30084906 DOI: 10.1093/aje/kwy145] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/09/2018] [Indexed: 01/08/2023] Open
Abstract
Despite all we have learned since 1918 about influenza virus and immunity, available influenza vaccines remain inadequate to control outbreaks of unexpected strains. Universal vaccines not requiring strain matching would be a major improvement. Their composition would be independent of predicting circulating viruses and thus potentially effective against unexpected drift or pandemic strains. This commentary explores progress with candidate universal vaccines based on various target antigens. Candidates include vaccines based on conserved viral proteins such as nucleoprotein and matrix, on the conserved hemagglutinin (HA) stem, and various combinations. Discussion covers the differing evidence for each candidate vaccine demonstrating protection in animals against influenza viruses of widely divergent HA subtypes and groups; durability of protection; routes of administration, including mucosal, providing local immunity; and reduction of transmission. Human trials of some candidate universal vaccines have been completed or are underway. Interestingly, the HA stem, like nucleoprotein and matrix, induces immunity that permits some virus replication and emergence of escape mutants fit enough to cause disease. Vaccination with multiple target antigens will thus have advantages over use of single antigens. Ultimately, a universal vaccine providing long-term protection against all influenza virus strains might contribute to pandemic control and routine vaccination.
Collapse
Affiliation(s)
- Suzanne L Epstein
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
31
|
McLean HQ, Caspard H, Griffin MR, Gaglani M, Peters TR, Poehling KA, Ambrose CS, Belongia EA. Association of Prior Vaccination With Influenza Vaccine Effectiveness in Children Receiving Live Attenuated or Inactivated Vaccine. JAMA Netw Open 2018; 1:e183742. [PMID: 30646262 PMCID: PMC6324442 DOI: 10.1001/jamanetworkopen.2018.3742] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IMPORTANCE Some studies have reported negative effects of prior-season influenza vaccination. Prior-season influenza vaccination effects on vaccine effectiveness (VE) in children are not well understood. OBJECTIVE To assess the association of prior-season influenza vaccination with subsequent VE in children aged 2 to 17 years. DESIGN, SETTING, AND PARTICIPANTS This multiseason, test-negative case-control study was conducted in outpatient clinics at 4 US sites among children aged 2 to 17 years with a medically attended febrile acute respiratory illness. Participants were recruited during the 2013-2014, 2014-2015, and 2015-2016 seasons when influenza circulated locally. Cases were children with influenza confirmed by reverse-transcription polymerase chain reaction. Test-negative control individuals were children with negative test results for influenza. EXPOSURES Vaccination history, including influenza vaccine type received in the enrollment season (live attenuated influenza vaccine [LAIV], inactivated influenza vaccine [IIV], or no vaccine) and season before enrollment (LAIV, IIV, or no vaccine), determined from medical records and immunization registries. MAIN OUTCOMES AND MEASURES LAIV and IIV effectiveness by influenza type and subtype (influenza A[H1N1]pdm09, influenza A[H3N2], or influenza B), estimated as 100 × (1 - odds ratio) in a logistic regression model with adjustment for potential confounders. Prior season vaccination associations were assessed with an interaction term. RESULTS Of 3369 children (1749 [52%] male; median age, 6.6 years [range, 2-17 years]) included in the analysis, 772 (23%) had a positive test result for influenza and 1674 (50%) were vaccinated in the enrollment season. Among LAIV recipients, VE against influenza A(H3N2) was higher among children vaccinated in both the enrollment and 1 prior season (50.3% [95% CI, 17.0% to 70.2%]) than among those without 1 prior season vaccination (-82.4% [95% CI, -267.5% to 9.5%], interaction P < .001). The effectiveness of LAIV against influenza A(H1N1)pdm09 was not associated with prior season vaccination among those with prior season vaccination (47.5% [95% CI, 11.4% to 68.9%]) and among those without prior season vaccination (7.8% [95% CI, -101.9% to 57.9%]) (interaction P = .37). Prior season vaccination was not associated with effectiveness of IIV against influenza A(H3N2) (38.7% [95% CI, 6.8% to 59.6%] among those with prior-season vaccination and 23.2% [95% CI, -38.3% to 57.4%] among those without prior-season vaccination, interaction P = .16) or with effectiveness of IIV against influenza A[H1N1]pdm09 (72.4% [95% CI, 56.0% to 82.7%] among those with prior season vaccination and 67.5% [95% CI, 32.1% to 84.4%] among those without prior season vaccination, interaction P = .93). Residual protection from prior season vaccination only (no vaccination in the enrollment season) was observed for influenza B (LAIV: 60.0% [95% CI, 36.8% to 74.7%]; IIV: 60.0% [36.9% to 74.6%]). Similar results were observed in analyses that included repeated vaccination in 2 and 3 prior seasons. CONCLUSIONS AND RELEVANCE Influenza VE varied by influenza type and subtype and vaccine type, but prior-season vaccination was not associated with reduced VE. These findings support current recommendations for annual influenza vaccination of children.
Collapse
Affiliation(s)
| | | | | | - Manjusha Gaglani
- Baylor Scott and White Health, Texas A&M University Health Science Center College of Medicine, Temple
| | | | | | | | | |
Collapse
|
32
|
Korenkov DA, Laurie KL, Reading PC, Carolan LA, Chan KF, Isakova-Sivak II, Smolonogina TA, Subbarao K, Barr IG, Villanueva J, Shcherbik S, Bousse T, Rudenko LG. Safety, immunogenicity and protection of A(H3N2) live attenuated influenza vaccines containing wild-type nucleoprotein in a ferret model. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2018; 64:95-104. [PMID: 29929009 PMCID: PMC6330673 DOI: 10.1016/j.meegid.2018.06.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 11/21/2022]
Abstract
Live attenuated influenza vaccines (LAIVs) are promising tools for the induction of broad protection from influenza due to their ability to stimulate cross-reactive T cells against influenza pathogens. One of the major targets for cytotoxic T-cell immunity is viral nucleoprotein (NP), which is relatively conserved among antigenically distant influenza viruses. Nevertheless, a diversity of epitope composition has been found in the NP protein of different lineages of influenza A viruses. The H2N2 master donor virus which is currently used as a backbone for the LAIV and donor of the six genomic segments encoding the internal proteins, A/Leningrad/134/17/57 (MDV Len/17), was isolated 60 years ago. As such, NP-specific T-cell immunity induced upon vaccination with classical LAIVs with a 6:2 genome composition containing this older NP might be suboptimal against currently circulating influenza viruses. In this study, a panel of H3N2 LAIV candidates with wild-type NP genes derived from circulating viruses were generated by reverse genetics (5:3 genome composition). These viruses displayed the cold adaptation and temperature sensitivity phenotypes of MDV Len/17 in vitro. LAIVs with both 6:2 and 5:3 genome compositions were attenuated and replicated to a similar extent in the upper respiratory tract of ferrets. LAIVs were immunogenic as high neutralizing and hemagglutination inhibition serum antibody titers were detected 21 days after infection. All vaccinated animals were protected against infection with heterologous H3N2 influenza A viruses. Thus, LAIV with a 5:3 genome composition is safe, immunogenic and can induce cross-protective immunity.
Collapse
MESH Headings
- Animal Diseases/immunology
- Animal Diseases/prevention & control
- Animal Diseases/virology
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Disease Models, Animal
- Female
- Ferrets
- Genome, Viral
- Immunogenicity, Vaccine
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/adverse effects
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Male
- Neutralization Tests
- Nucleoproteins/genetics
- Nucleoproteins/immunology
- Orthomyxoviridae Infections/veterinary
- Vaccination
- Vaccines, Attenuated/adverse effects
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
Collapse
Affiliation(s)
- Daniil A Korenkov
- Department of Virology, Institute of Experimental Medicine, 12, acad. Pavlova street, Saint Petersburg, Russia.
| | - Karen L Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at The Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Patrick C Reading
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at The Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Louise A Carolan
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at The Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Kok Fei Chan
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at The Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Irina I Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 12, acad. Pavlova street, Saint Petersburg, Russia
| | - Tatiana A Smolonogina
- Department of Virology, Institute of Experimental Medicine, 12, acad. Pavlova street, Saint Petersburg, Russia
| | - Kanta Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at The Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at The Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | | | | | - Tatiana Bousse
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Larisa G Rudenko
- Department of Virology, Institute of Experimental Medicine, 12, acad. Pavlova street, Saint Petersburg, Russia
| |
Collapse
|
33
|
Kotomina T, Korenkov D, Matyushenko V, Prokopenko P, Rudenko L, Isakova-Sivak I. Live attenuated influenza vaccine viral vector induces functional cytotoxic T-cell immune response against foreign CD8+ T-cell epitopes inserted into NA and NS1 genes using the 2A self-cleavage site. Hum Vaccin Immunother 2018; 14:2964-2970. [PMID: 30024831 DOI: 10.1080/21645515.2018.1502529] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The development of viral vector vaccines against various pathogens for which conventional vaccination approaches are not applicable has been a priority for a number of years. One promising approach is the insertion of immunodominant conservative cytotoxic T-cell (CTL) epitopes into the genome of a viral vector, which then delivers these epitopes to target cells, inducing immunity. Many different viruses have been assessed as viral vectors for CTL-based vaccines, but only a few of them are clinically relevant, mainly because of safety issues and limited knowledge about their performance in humans. In this regard, the use of licensed cold-adapted live attenuated influenza vaccine (LAIV) viruses as a vector delivery system has clear advantages for CTL-based vector vaccines against other respiratory pathogens: LAIV is known to induce all arms of the adaptive immune system and is administered via nasal spray, and its production process is relatively easy and inexpensive. Here we present the first results of the use of an LAIV backbone for designing a CTL epitope-based vaccine against respiratory syncytial virus (RSV). The chimeric LAIV-RSV vaccine candidates were attenuated in mice and induced strong, fully functional CTL immunity in this animal model.
Collapse
Affiliation(s)
- Tatiana Kotomina
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - Daniil Korenkov
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - Victoria Matyushenko
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - Polina Prokopenko
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - Larisa Rudenko
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - Irina Isakova-Sivak
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| |
Collapse
|
34
|
Zhou F, Trieu MC, Davies R, Cox RJ. Improving influenza vaccines: challenges to effective implementation. Curr Opin Immunol 2018; 53:88-95. [DOI: 10.1016/j.coi.2018.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/08/2018] [Accepted: 04/13/2018] [Indexed: 12/15/2022]
|
35
|
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
|
36
|
Mohn KGI, Zhou F. Clinical Expectations for Better Influenza Virus Vaccines-Perspectives from the Young Investigators' Point of View. Vaccines (Basel) 2018; 6:E32. [PMID: 29861454 PMCID: PMC6027204 DOI: 10.3390/vaccines6020032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/20/2023] Open
Abstract
The influenza virus is one of a few viruses that is capable of rendering an otherwise healthy person acutly bedridden for several days. This impressive knock-out effect, without prodromal symptoms, challenges our immune system. The influenza virus undergoes continuous mutations, escaping our pre-existing immunity and causing epidemics, and its segmented genome is subject to reassortment, resulting in novel viruses with pandemic potential. The personal and socieoeconomic burden from influenza is high. Vaccination is the most cost-effective countermeasure, with several vaccines that are available. The current limitations in vaccine effectivness, combined with the need for yearly updating of vaccine strains, is a driving force for research into developing new and improved influenza vaccines. The lack of public concern about influenza severity, and misleading information concerning vaccine safety contribute to low vaccination coverage even in high-risk groups. The success of future influeza vaccines will depend on an increased public awarness of the disease, and hence, the need for vaccination-aided through improved rapid diagnositics. The vaccines must be safe and broadly acting, with new, measurable correlates of protection and robust post-marketing safety studies, to improve the confidence in influenza vaccines.
Collapse
Affiliation(s)
- Kristin G-I Mohn
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
- Emergency Care clinic, Haukeland University Hospital, Bergen 5021, Norway.
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
- K.G. Jebsen Centre for Influenza Vaccine Research, Department of Clinical Science, University of Bergen, Bergen 5021, Norway.
| |
Collapse
|
37
|
Pusch E, Renz H, Skevaki C. Respiratory virus-induced heterologous immunity: Part of the problem or part of the solution? ALLERGO JOURNAL 2018; 27:28-45. [PMID: 32300267 PMCID: PMC7149200 DOI: 10.1007/s15007-018-1580-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/15/2018] [Indexed: 12/31/2022]
Abstract
Purpose To provide current knowledge on respiratory virus-induced heterologous immunity (HI) with a focus on humoral and cellular cross-reactivity. Adaptive heterologous immune responses have broad implications on infection, autoimmunity, allergy and transplant immunology. A better understanding of the mechanisms involved might ultimately open up possibilities for disease prevention, for example by vaccination. Methods A structured literature search was performed using Medline and PubMed to provide an overview of the current knowledge on respiratory-virus induced adaptive HI. Results In HI the immune response towards one antigen results in an alteration of the immune response towards a second antigen. We provide an overview of respiratory virus-induced HI, including viruses such as respiratory syncytial virus (RSV), rhinovirus (RV), coronavirus (CoV) and influenza virus (IV). We discuss T cell receptor (TCR) and humoral cross-reactivity as mechanisms of HI involving those respiratory viruses. Topics covered include HI between respiratory viruses as well as between respiratory viruses and other pathogens. Newly developed vaccines, which have the potential to provide protection against multiple virus strains are also discussed. Furthermore, respiratory viruses have been implicated in the development of autoimmune diseases, such as narcolepsy, Guillain-Barré syndrome, type 1 diabetes or myocarditis. Finally, we discuss the role of respiratory viruses in asthma and the hygiene hypothesis, and review our recent findings on HI between IV and allergens, which leads to protection from experimental asthma. Conclusion Respiratory-virus induced HI may have protective but also detrimental effects on the host. Respiratory viral infections contribute to asthma or autoimmune disease development, but on the other hand, a lack of microbial encounter is associated with an increasing number of allergic as well as autoimmune diseases. Future research might help identify the elements which determine a protective or detrimental outcome in HI-based mechanisms.
Collapse
Affiliation(s)
- Emanuel Pusch
- Institute of Laboratory Medicine, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| |
Collapse
|
38
|
Intranasal Live Influenza Vaccine Priming Elicits Localized B Cell Responses in Mediastinal Lymph Nodes. J Virol 2018; 92:JVI.01970-17. [PMID: 29444938 DOI: 10.1128/jvi.01970-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/26/2018] [Indexed: 01/05/2023] Open
Abstract
Pandemic live attenuated influenza vaccines (pLAIV) prime subjects for a robust neutralizing antibody response upon subsequent administration of a pandemic inactivated subunit vaccine (pISV). However, a difference was not detected in H5-specific memory B cells in the peripheral blood between pLAIV-primed and unprimed subjects prior to pISV boost. To investigate the mechanism underlying pLAIV priming, we vaccinated groups of 12 African green monkeys (AGMs) with H5N1 pISV or pLAIV alone or H5N1 pLAIV followed by pISV and examined immunity systemically and in local draining lymph nodes (LN). The AGM model recapitulated the serologic observations from clinical studies. Interestingly, H5N1 pLAIV induced robust germinal center B cell responses in the mediastinal LN (MLN). Subsequent boosting with H5N1 pISV drove increases in H5-specific B cells in the axillary LN, spleen, and circulation in H5N1 pLAIV-primed animals. Thus, H5N1 pLAIV primes localized B cell responses in the MLN that are recalled systemically following pISV boost. These data provide mechanistic insights for the generation of robust humoral responses via prime-boost vaccination.IMPORTANCE We have previously shown that pandemic live attenuated influenza vaccines (pLAIV) prime for a rapid and robust antibody response on subsequent administration of inactivated subunit vaccine (pISV). This is observed even in individuals who had undetectable antibody (Ab) responses following the initial vaccination. To define the mechanistic basis of pLAIV priming, we turned to a nonhuman primate model and performed a detailed analysis of B cell responses in systemic and local lymphoid tissues following prime-boost vaccination with pLAIV and pISV. We show that the nonhuman primate model recapitulates the serologic observations from clinical studies. Further, we found that pLAIVs induced robust germinal center B cell responses in the mediastinal lymph node. Subsequent boosting with pISV in pLAIV-primed animals resulted in detection of B cells in the axillary lymph nodes, spleen, and peripheral blood. We demonstrate that intranasally administered pLAIV elicits a highly localized germinal center B cell response in the mediastinal lymph node that is rapidly recalled following pISV boost into germinal center reactions at numerous distant immune sites.
Collapse
|
39
|
Clemens EB, van de Sandt C, Wong SS, Wakim LM, Valkenburg SA. Harnessing the Power of T Cells: The Promising Hope for a Universal Influenza Vaccine. Vaccines (Basel) 2018; 6:vaccines6020018. [PMID: 29587436 PMCID: PMC6027237 DOI: 10.3390/vaccines6020018] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Next-generation vaccines that utilize T cells could potentially overcome the limitations of current influenza vaccines that rely on antibodies to provide narrow subtype-specific protection and are prone to antigenic mismatch with circulating strains. Evidence from animal models shows that T cells can provide heterosubtypic protection and are crucial for immune control of influenza virus infections. This has provided hope for the design of a universal vaccine able to prime against diverse influenza virus strains and subtypes. However, multiple hurdles exist for the realisation of a universal T cell vaccine. Overall primary concerns are: extrapolating human clinical studies, seeding durable effective T cell resident memory (Trm), population human leucocyte antigen (HLA) coverage, and the potential for T cell-mediated immune escape. Further comprehensive human clinical data is needed during natural infection to validate the protective role T cells play during infection in the absence of antibodies. Furthermore, fundamental questions still exist regarding the site, longevity and duration, quantity, and phenotype of T cells needed for optimal protection. Standardised experimental methods, and eventually simplified commercial assays, to assess peripheral influenza-specific T cell responses are needed for larger-scale clinical studies of T cells as a correlate of protection against influenza infection. The design and implementation of a T cell-inducing vaccine will require a consensus on the level of protection acceptable in the community, which may not provide sterilizing immunity but could protect the individual from severe disease, reduce the length of infection, and potentially reduce transmission in the community. Therefore, increasing the standard of care potentially offered by T cell vaccines should be considered in the context of pandemic preparedness and zoonotic infections, and in combination with improved antibody vaccine targeting methods. Current pandemic vaccine preparedness measures and ongoing clinical trials under-utilise T cell-inducing vaccines, reflecting the myriad questions that remain about how, when, where, and which T cells are needed to fight influenza virus infection. This review aims to bring together basic fundamentals of T cell biology with human clinical data, which need to be considered for the implementation of a universal vaccine against influenza that harnesses the power of T cells.
Collapse
Affiliation(s)
- E Bridie Clemens
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Carolien van de Sandt
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Sook San Wong
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Sophie A Valkenburg
- HKU Pasteur Research Pole, School of Public Health, University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
40
|
Pusch E, Renz H, Skevaki C. Respiratory virus-induced heterologous immunity: Part of the problem or part of the solution? ACTA ACUST UNITED AC 2018; 27:79-96. [PMID: 32226720 PMCID: PMC7100437 DOI: 10.1007/s40629-018-0056-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/15/2018] [Indexed: 12/13/2022]
Abstract
Purpose To provide current knowledge on respiratory virus-induced heterologous immunity (HI) with a focus on humoral and cellular cross-reactivity. Adaptive heterologous immune responses have broad implications on infection, autoimmunity, allergy and transplant immunology. A better understanding of the mechanisms involved might ultimately open up possibilities for disease prevention, for example by vaccination. Methods A structured literature search was performed using Medline and PubMed to provide an overview of the current knowledge on respiratory-virus induced adaptive HI. Results In HI the immune response towards one antigen results in an alteration of the immune response towards a second antigen. We provide an overview of respiratory virus-induced HI, including viruses such as respiratory syncytial virus (RSV), rhinovirus (RV), coronavirus (CoV) and influenza virus (IV). We discuss T cell receptor (TCR) and humoral cross-reactivity as mechanisms of HI involving those respiratory viruses. Topics covered include HI between respiratory viruses as well as between respiratory viruses and other pathogens. Newly developed vaccines which have the potential to provide protection against multiple virus strains are also discussed. Furthermore, respiratory viruses have been implicated in the development of autoimmune diseases, such as narcolepsy, Guillain–Barré syndrome, type 1 diabetes or myocarditis. Finally, we discuss the role of respiratory viruses in asthma and the hygiene hypothesis, and review our recent findings on HI between IV and allergens, which leads to protection from experimental asthma. Conclusion Respiratory-virus induced HI may have protective but also detrimental effects on the host. Respiratory viral infections contribute to asthma or autoimmune disease development, but on the other hand, a lack of microbial encounter is associated with an increasing number of allergic as well as autoimmune diseases. Future research might help identify the elements which determine a protective or detrimental outcome in HI-based mechanisms.
Collapse
Affiliation(s)
- Emanuel Pusch
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine and Pathobiochemistry, Member of the German Center for Lung Research (DZL), Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| |
Collapse
|
41
|
Kumar A, Meldgaard TS, Bertholet S. Novel Platforms for the Development of a Universal Influenza Vaccine. Front Immunol 2018; 9:600. [PMID: 29628926 PMCID: PMC5877485 DOI: 10.3389/fimmu.2018.00600] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/09/2018] [Indexed: 12/19/2022] Open
Abstract
Despite advancements in immunotherapeutic approaches, influenza continues to cause severe illness, particularly among immunocompromised individuals, young children, and elderly adults. Vaccination is the most effective way to reduce rates of morbidity and mortality caused by influenza viruses. Frequent genetic shift and drift among influenza-virus strains with the resultant disparity between circulating and vaccine virus strains limits the effectiveness of the available conventional influenza vaccines. One approach to overcome this limitation is to develop a universal influenza vaccine that could provide protection against all subtypes of influenza viruses. Moreover, the development of a novel or improved universal influenza vaccines may be greatly facilitated by new technologies including virus-like particles, T-cell-inducing peptides and recombinant proteins, synthetic viruses, broadly neutralizing antibodies, and nucleic acid-based vaccines. This review discusses recent scientific advances in the development of next-generation universal influenza vaccines.
Collapse
Affiliation(s)
- Arun Kumar
- GSK, Research and Development Center, Siena, Italy.,Linköping University, Linköping, Sweden
| | - Trine Sundebo Meldgaard
- GSK, Research and Development Center, Siena, Italy.,DTU Nanotech, Technical University of Denmark, Copenhagen, Denmark
| | - Sylvie Bertholet
- GSK, Research and Development Center, Siena, Italy.,GSK, Research and Development Center, Rockville, MD, United States
| |
Collapse
|
42
|
Gallinaro A, Borghi M, Bona R, Grasso F, Calzoletti L, Palladino L, Cecchetti S, Vescio MF, Macchia D, Morante V, Canitano A, Temperton N, Castrucci MR, Salvatore M, Michelini Z, Cara A, Negri D. Integrase Defective Lentiviral Vector as a Vaccine Platform for Delivering Influenza Antigens. Front Immunol 2018; 9:171. [PMID: 29459873 PMCID: PMC5807328 DOI: 10.3389/fimmu.2018.00171] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 01/19/2018] [Indexed: 12/31/2022] Open
Abstract
Viral vectors represent an attractive technology for vaccine delivery. We exploited the integrase defective lentiviral vector (IDLV) as a platform for delivering relevant antigens within the context of the ADITEC collaborative research program. In particular, Influenza virus hemagglutinin (HA) and nucleoprotein (NP) were delivered by IDLVs while H1N1 A/California/7/2009 subunit vaccine (HAp) with or without adjuvant was used to compare the immune response in a murine model of immunization. In order to maximize the antibody response against HA, both IDLVs were also pseudotyped with HA (IDLV-HA/HA and IDLV-NP/HA, respectively). Groups of CB6F1 mice were immunized intramuscularly with a single dose of IDLV-NP/HA, IDLV-HA/HA, HAp alone, or with HAp together with the systemic adjuvant MF59. Six months after the vaccine prime all groups were boosted with HAp alone. Cellular and antibody responses to influenza antigens were measured at different time points after the immunizations. Mice immunized with HA-pseudotyped IDLVs showed similar levels of anti-H1N1 IgG over time, evaluated by ELISA, which were comparable to those induced by HAp + MF59 vaccination, but significantly higher than those induced by HAp alone. The boost with HAp alone induced an increase of antibodies in all groups, and the responses were maintained at higher levels up to 18 weeks post-boost. The antibody response was functional and persistent overtime, capable of neutralizing virus infectivity, as evaluated by hemagglutination inhibition and microneutralization assays. Moreover, since neuraminidase (NA)-expressing plasmid was included during IDLV preparation, immunization with IDLV-NP/HA and IDLV-HA/HA also induced functional anti-NA antibodies, evaluated by enzyme-linked lectin assay. IFNγ-ELISPOT showed evidence of HA-specific response in IDLV-HA/HA immunized animals and persistent NP-specific CD8+ T cell response in IDLV-NP/HA immunized mice. Taken together our results indicate that IDLV can be harnessed for producing a vaccine able to induce a comprehensive immune response, including functional antibodies directed toward HA and NA proteins present on the vector particles in addition to a functional T cell response directed to the protein transcribed from the vector.
Collapse
Affiliation(s)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta Bona
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Felicia Grasso
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Calzoletti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Serena Cecchetti
- Confocal Microscopy Unit NMR, Confocal Microscopy Area Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Daniele Macchia
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Valeria Morante
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Canitano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Kent, United Kingdom
| | | | - Mirella Salvatore
- Department of Medicine, Weill Cornell Medical College, New York, United States
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
43
|
Korenkov D, Nguyen THO, Isakova-Sivak I, Smolonogina T, Brown LE, Kedzierska K, Rudenko L. Live Attenuated Influenza Vaccines engineered to express the nucleoprotein of a recent isolate stimulate human influenza CD8 + T cells more relevant to current infections. Hum Vaccin Immunother 2018; 14:941-946. [PMID: 29252117 DOI: 10.1080/21645515.2017.1417713] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Live attenuated influenza vaccines (LAIV) induce CD8+ T lymphocyte responses that play an important role in killing virus-infected cells. Despite the relative conservation of internal influenza A proteins, the epitopes recognized by T cells can undergo drift under immune pressure. The internal proteins of Russian LAIVs are derived from the master donor virus A/Leningrad/134/17/57 (Len/17) isolated 60 years ago and as such, some CD8+ T cell epitopes may vary between the vaccine and circulating wild-type strains. To partially overcome this issue, the nucleoprotein (NP) gene of wild-type virus can be incorporated into LAIV reassortant virus, along with the HA and NA genes. The present study compares the human CD8+ T cell memory responses to H3N2 LAIVs with the Len/17 or the wild-type NP using an in vitro model.
Collapse
Affiliation(s)
- D Korenkov
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia.,b Department of Microbiology & Immunology , University of Melbourne, at The Peter Doherty Institute for Infection & Immunity , Melbourne , VIC , Australia
| | - T H O Nguyen
- b Department of Microbiology & Immunology , University of Melbourne, at The Peter Doherty Institute for Infection & Immunity , Melbourne , VIC , Australia
| | - I Isakova-Sivak
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - T Smolonogina
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| | - L E Brown
- b Department of Microbiology & Immunology , University of Melbourne, at The Peter Doherty Institute for Infection & Immunity , Melbourne , VIC , Australia
| | - K Kedzierska
- b Department of Microbiology & Immunology , University of Melbourne, at The Peter Doherty Institute for Infection & Immunity , Melbourne , VIC , Australia
| | - L Rudenko
- a Department of Virology , Institute of Experimental Medicine , Saint Petersburg , Russia
| |
Collapse
|
44
|
Gill MA, Schlaudecker EP. Perspectives from the Society for Pediatric Research: Decreased Effectiveness of the Live Attenuated Influenza Vaccine. Pediatr Res 2018; 83:31-40. [PMID: 28945700 DOI: 10.1038/pr.2017.239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/15/2017] [Indexed: 11/09/2022]
Abstract
The intranasal live attenuated influenza vaccine (LAIV), FluMist, has been widely appreciated by pediatricians, parents, and children alike for its ease of administration. However, concerns regarding lack of effectiveness in recent influenza seasons led to the CDC Advisory Committee on Immunization Practices (ACIP) recommendation to administer inactivated influenza vaccines (IIVs), and not LAIV, during the 2016-17 and 2017-18 seasons. Given that data from previous years demonstrated equivalent and even improved efficacy of LAIV compared with IIV, these recent data were surprising, raising many questions about the potential mechanisms underlying this change. This review seeks to summarize the history of LAIV studies and ACIP recommendations with a focus on the recent decrease in vaccine effectiveness (VE) and discordant results among studies performed in different countries. Decreased VE for A/H1N1pdm09 viruses represents the most consistent finding across studies, as VE has been low every season these viruses predominated since 2010-11. Potential explanations underlying diminished effectiveness include the hypothesis that prior vaccination, reduced thermostability of A/H1N1pdm09, addition of a fourth virus, or reduced replication fitness of A/H1N1pdm09 strains may have contributed to this phenomenon. Ongoing studies and potential alterations to LAIV formulations provide hope for a return of effective LAIV in future influenza seasons.
Collapse
Affiliation(s)
- Michelle A Gill
- Division of Infectious Diseases, Departments of Pediatrics, Internal Medicine, and Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Elizabeth P Schlaudecker
- Division of Infectious Diseases, Global Health Center, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|