1
|
Russell CA, Fouchier RAM, Ghaswalla P, Park Y, Vicic N, Ananworanich J, Nachbagauer R, Rudin D. Seasonal influenza vaccine performance and the potential benefits of mRNA vaccines. Hum Vaccin Immunother 2024; 20:2336357. [PMID: 38619079 PMCID: PMC11020595 DOI: 10.1080/21645515.2024.2336357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Influenza remains a public health threat, partly due to suboptimal effectiveness of vaccines. One factor impacting vaccine effectiveness is strain mismatch, occurring when vaccines no longer match circulating strains due to antigenic drift or the incorporation of inadvertent (eg, egg-adaptive) mutations during vaccine manufacturing. In this review, we summarize the evidence for antigenic drift of circulating viruses and/or egg-adaptive mutations occurring in vaccine strains during the 2011-2020 influenza seasons. Evidence suggests that antigenic drift led to vaccine mismatch during four seasons and that egg-adaptive mutations caused vaccine mismatch during six seasons. These findings highlight the need for alternative vaccine development platforms. Recently, vaccines based on mRNA technology have demonstrated efficacy against SARS-CoV-2 and respiratory syncytial virus and are under clinical evaluation for seasonal influenza. We discuss the potential for mRNA vaccines to address strain mismatch, as well as new multi-component strategies using the mRNA platform to improve vaccine effectiveness.
Collapse
Affiliation(s)
- Colin A. Russell
- Department of Medical Microbiology & Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Ron A. M. Fouchier
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
2
|
Harada Y, Takahashi H, Fujimoto T, Horikoshi F, Chida S, Tanaka K, Minari K, Tanimoto Y, Fujisaki S, Miura H, Nakauchi M, Shimasaki N, Suzuki Y, Arita T, Hamamoto I, Yamamoto N, Hasegawa H, Odagiri T, Tashiro M, Nobusawa E. Evaluation of a qualified MDCK cell line for virus isolation to develop cell-based influenza vaccine viruses with appropriate antigenicity. Vaccine 2024; 42:126242. [PMID: 39213922 DOI: 10.1016/j.vaccine.2024.126242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
We established a qualified Madin-Darby canine kidney cell line (qMDCK-Cs) and investigated its suitability for source virus isolation to develop cell-based seasonal influenza vaccine viruses using vaccine manufacturer cells (Manuf-Cs). When inoculated with 81 influenza-positive clinical specimens, the initial virus isolation efficiency of qMDCK-Cs was exceeded 70%. Among the qMDCK-C isolates, 100% of the A/H1N1pdm09, B/Victoria and B/Yamagata strains and >70% of the A/H3N2 strains showed antigenicity equivalent to that of the contemporary vaccine or relevant viruses in haemagglutination inhibition (HI) or virus neutralization (VN) tests using ferret antisera. These qMDCK-C isolates were propagated in Manuf-Cs (MDCK and Vero cells) (Manuf-C viruses) to develop vaccine viruses. In reciprocal antigenicity tests, ferret antisera raised against corresponding reference viruses and Manuf-C viruses recognized 29 of 31 Manuf-C viruses and corresponding reference viruses, respectively at HI or VN titres more than half of the homologous virus titres, which is the antigenicity criterion for cell culture seasonal influenza vaccine viruses specified by the World Health Organization. Furthermore, ferret antisera against these Manuf-C viruses recognized ≥95% of the viruses circulating during the relevant influenza season with HI or VN titres greater than one-quarter of the homologous virus titres. No cell line-specific amino acid substitutions were observed in the resulting viruses. However, polymorphisms at positions 158/160 of H3HA, 148/151 of N2NA and 197/199 of B/Victoria HA were occasionally detected in the qMDCK-C and Manuf-C viruses but barely affected the viral antigenicity. These results indicated that qMDCK-Cs are suitable for isolating influenza viruses that can serve as a source of antigenically appropriate vaccine viruses. The use of the qMDCK-C isolates will eliminates the need for clinical sample collection, virus isolation, and antigenicity analysis every season, and is expected to contribute to the promotion of vaccine virus development using manufacturer cells.
Collapse
Affiliation(s)
- Yuichi Harada
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan; Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Hitoshi Takahashi
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Takao Fujimoto
- BIKEN CO., Ltd., 4-1-70, Seto-Cho, Kan-Onji, Kagawa 768-0065, Japan
| | | | - Shuhei Chida
- BIKEN CO., Ltd., 4-1-70, Seto-Cho, Kan-Onji, Kagawa 768-0065, Japan
| | - Kenji Tanaka
- Daiichi Sankyo Biotech Co., Ltd., 6-111 Arai, Kitamoto City, Saitama Prefecture 364-0026, Japan
| | - Kenji Minari
- Takeda Pharmaceutical Company Limited, Hikari Plant, 4720 Takeda, Mitsui, Hikari City, Yamaguchi 743-8502, Japan
| | - Yoshimi Tanimoto
- Takeda Pharmaceutical Company Limited, Hikari Plant, 4720 Takeda, Mitsui, Hikari City, Yamaguchi 743-8502, Japan
| | - Seiichiro Fujisaki
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Hideka Miura
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Mina Nakauchi
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Noriko Shimasaki
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan; Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Yasushi Suzuki
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Tomoko Arita
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Itsuki Hamamoto
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Norio Yamamoto
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Hideki Hasegawa
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Takato Odagiri
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Masato Tashiro
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan
| | - Eri Nobusawa
- Research Center for Influenza and Respiratory Viruses, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama, Tokyo 208-0011, Japan.
| |
Collapse
|
3
|
Clark TW, Tregoning JS, Lister H, Poletti T, Amin F, Nguyen-Van-Tam JS. Recent advances in the influenza virus vaccine landscape: a comprehensive overview of technologies and trials. Clin Microbiol Rev 2024:e0002524. [PMID: 39360831 DOI: 10.1128/cmr.00025-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
SUMMARYIn the United Kingdom (UK) in 2022/23, influenza virus infections returned to the levels recorded before the COVID-19 pandemic, exerting a substantial burden on an already stretched National Health Service (NHS) through increased primary and emergency care visits and subsequent hospitalizations. Population groups ≤4 years and ≥65 years of age, and those with underlying health conditions, are at the greatest risk of influenza-related hospitalization. Recent advances in influenza virus vaccine technologies may help to mitigate this burden. This review aims to summarize advances in the influenza virus vaccine landscape by describing the different technologies that are currently in use in the UK and more widely. The review also describes vaccine technologies that are under development, including mRNA, and universal influenza virus vaccines which aim to provide broader or increased protection. This is an exciting and important era for influenza virus vaccinations, and advances are critical to protect against a disease that still exerts a substantial burden across all populations and disproportionately impacts the most vulnerable, despite it being over 80 years since the first influenza virus vaccines were deployed.
Collapse
Affiliation(s)
- Tristan W Clark
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
4
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
5
|
Hsu D, Jayaraman A, Pucci A, Joshi R, Mancini K, Chen HL, Koslovsky K, Mao X, Choi A, Henry C, Vakil J, Stadlbauer D, Jorquera P, Arunkumar GA, Sanchez-Crespo NE, Wadsworth LT, Bhupathy V, Du E, Avanesov A, Ananworanich J, Nachbagauer R. Safety and immunogenicity of mRNA-based seasonal influenza vaccines formulated to include multiple A/H3N2 strains with or without the B/Yamagata strain in US adults aged 50-75 years: a phase 1/2, open-label, randomised trial. THE LANCET. INFECTIOUS DISEASES 2024:S1473-3099(24)00493-6. [PMID: 39245055 DOI: 10.1016/s1473-3099(24)00493-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Inclusion of additional influenza A/H3N2 strains in seasonal influenza vaccines could expand coverage against multiple, antigenically distinct, cocirculating A/H3N2 clades and potentially replace the no longer circulating B/Yamagata strain. We aimed to evaluate the safety and immunogenicity of three next-generation seasonal influenza mRNA vaccines with different compositions that encode for haemagglutinins of multiple A/H3N2 strains, with or without the B/Yamagata strain, in adults. METHODS This randomised, open-label, phase 1/2 trial enrolled healthy adults aged 50-75 years across 22 sites in the USA. Participants were randomly assigned (1:1:1:1:1:1:1) via interactive response technology to receive a single dose of mRNA-1011.1 (pentavalent; containing one additional A/H3N2 strain [Newcastle]), mRNA-1011.2 (quadrivalent; B/Yamagata replaced with one additional A/H3N2 strain [Newcastle]), mRNA-1012 at one of two dose levels (pentavalent; B/Yamagata replaced with two additional A/H3N2 strains [Newcastle and Hong Kong]), or one of three quadrivalent mRNA-1010 controls each encoding one of the A/H3N2 study strains. The primary outcomes were safety, evaluated in all randomly assigned participants who received a study vaccination (safety population), and reactogenicity, evaluated in all participants from the safety population who contributed any solicited adverse reaction data (solicited safety population). The secondary outcome was humoral immunogenicity of investigational mRNA vaccines at day 29 versus mRNA-1010 control vaccines based on haemagglutination inhibition antibody (HAI) assay in the per-protocol population. Here, we summarise findings from the planned interim analysis after participants had completed day 29. The study is registered with ClinicalTrials.gov, NCT05827068, and is ongoing. FINDINGS Between March 27 and May 9, 2023, 1183 participants were screened for eligibility, 699 (59·1%) were randomly assigned, and 696 (58·8%) received vaccination (safety population, n=696; solicited safety population, n=694; per-protocol population, n=646). 382 (55%) of the 696 participants in the safety population self-reported as female and 314 (45%) as male. Frequencies of solicited adverse reactions were similar across vaccine groups; 551 (79%) of 694 participants reported at least one solicited adverse reaction within 7 days after vaccination and 83 (12%) of 696 participants reported at least one unsolicited adverse event within 28 days after vaccination. No vaccine-related serious adverse events or deaths were reported. All three next-generation influenza vaccines elicited robust antibody responses against vaccine-matched influenza A and B strains at day 29 that were generally similar to mRNA-1010 controls, and higher responses against additional A/H3N2 strains that were not included within respective mRNA-1010 controls. Day 29 geometric mean fold rises in HAI titres from day 1 against vaccine-matched A/H3N2 strains were 3·0 (95% CI 2·6-3·6; Darwin) and 3·1 (2·6-3·8; Newcastle) for mRNA-1011.1; 3·3 (2·7-4·1; Darwin) and 4·2 (3·4-5·2; Newcastle) for mRNA-1011.2; 3·4 (2·9-4·0; Darwin), 4·5 (3·6-5·5; Newcastle), and 5·1 (4·2-6·2; Hong Kong) for mRNA-1012 50·0 μg; and 2·6 (2·2-3·1; Darwin), 3·7 (3·0-4·6; Newcastle), and 4·1 (3·3-5·1; Hong Kong) for mRNA-1012 62·5 μg. Inclusion of additional A/H3N2 strains did not reduce responses against influenza A/H1N1 or influenza B strains, and removal of B/Yamagata did not affect responses to B/Victoria. INTERPRETATION These data support the continued clinical development of mRNA-based next-generation seasonal influenza vaccines with broadened influenza A/H3N2 strain coverage. FUNDING Moderna.
Collapse
|
6
|
Yang F, Zhao X, Huo C, Miao X, Qin T, Chen S, Peng D, Liu X. An avian-origin internal backbone effectively increases the H5 subtype avian influenza vaccine candidate yield in both chicken embryonated eggs and MDCK cells. Poult Sci 2024; 103:103988. [PMID: 38970848 PMCID: PMC11269899 DOI: 10.1016/j.psj.2024.103988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/29/2024] [Accepted: 06/12/2024] [Indexed: 07/08/2024] Open
Abstract
Inactivated vaccines play an important role in preventing and controlling the epidemic caused by the H5 subtype avian influenza virus. The vaccine strains are updated in response to alterations in surface protein antigens, while an avian-derived vaccine internal backbone with a high replicative capacity in chicken embryonated eggs and MDCK cells is essential for vaccine development. In this study, we constructed recombinant viruses using the clade 2.3.4.4d A/chicken/Jiangsu/GY5/2017(H5N6, CkG) strain as the surface protein donor and the clade 2.3.4.4b A/duck/Jiangsu/84512/2017(H5N6, Dk8) strain with high replicative ability as an internal donor. After optimization, the integration of the M gene from the CkG into the internal genes from Dk8 (8GM) was selected as the high-yield vaccine internal backbone, as the combination improved the hemagglutinin1/nucleoprotein (HA1/NP) ratio in recombinant viruses. The r8GMΔG with attenuated hemagglutinin and neuraminidase from the CkG exhibited high-growth capacity in both chicken embryos and MDCK cell cultures. The inactivated r8GMΔG vaccine candidate also induced a higher hemagglutination inhibition antibody titer and microneutralization titer than the vaccine strain using PR8 as the internal backbone. Further, the inactivated r8GMΔG vaccine candidate provided complete protection against wild-type strain challenge. Therefore, our study provides a high-yield, easy-to-cultivate candidate donor as an internal gene backbone for vaccine development.
Collapse
Affiliation(s)
- Fan Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xinyu Zhao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Chenzhi Huo
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Xinyu Miao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 225009, China; The International Joint Laboratory for Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu 225009, China
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 225009, China; The International Joint Laboratory for Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu 225009, China
| | - Sujuan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 225009, China; The International Joint Laboratory for Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu 225009, China
| | - Daxin Peng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 225009, China; The International Joint Laboratory for Cooperation in Agriculture and Agricultural Product Safety, Ministry of Education, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu 225009, China.
| | - Xiufan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu 225009, China; Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
7
|
Cai M, Le Y, Gong Z, Dong T, Liu B, Su M, Li X, Peng F, Li Q, Nian X, Yu H, Wu Z, Zhang Z, Zhang J. Production, Passaging Stability, and Histological Analysis of Madin-Darby Canine Kidney Cells Cultured in a Low-Serum Medium. Vaccines (Basel) 2024; 12:991. [PMID: 39340023 PMCID: PMC11435615 DOI: 10.3390/vaccines12090991] [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: 07/04/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Madin-Darby canine kidney (MDCK) cells are commonly used to produce cell-based influenza vaccines. However, the role of the low-serum medium on the proliferation of MDCK cells and the propagation of the influenza virus has not been well studied. In the present study, we used 5 of 15 culture methods with different concentrations of a mixed medium and neonatal bovine serum (NBS) to determine the best culture medium. We found that a VP:M199 ratio of 1:2 (3% NBS) was suitable for culturing MDCK cells. Furthermore, the stable growth of MDCK cells and the production of the influenza virus were evaluated over long-term passaging. We found no significant difference in terms of cell growth and virus production between high and low passages of MDCK cells under low-serum culture conditions, regardless of influenza virus infection. Lastly, we performed a comparison of the transcriptomics and proteomics of MDCK cells cultured in VP:M199 = 1:2 (3% NBS) with those cultured in VP:M199 = 1:2 (5% NBS) before and after influenza virus infection. The transcriptome analysis showed that differentially expressed genes were predominantly enriched in the metabolic pathway and MAPK signaling pathway, indicating an activated state. This suggests that decreasing the concentration of serum in the medium from 5% to 3% may increase the metabolic activity of cells. Proteomics analysis showed that only a small number of differentially expressed proteins could not be enriched for analysis, indicating minimal difference in the protein levels of MDCK cells when the serum concentration in the medium was decreased from 5% to 3%. Altogether, our findings suggest that the screening and application of a low-serum medium provide a background for the development and optimization of cell-based influenza vaccines.
Collapse
Affiliation(s)
- Ming Cai
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Yang Le
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zheng Gong
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Tianbao Dong
- Center for Drug Evaluation and Inspection of HMPA (Hubei Center for Vaccine Inspection), Wuhan 430207, China
| | - Bo Liu
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Minne Su
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuedan Li
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Feixia Peng
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Qingda Li
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuanxuan Nian
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Hao Yu
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zheng Wu
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zhegang Zhang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Jiayou Zhang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| |
Collapse
|
8
|
Shahri MS, Sadeghi S, Hazegh Fetratjoo D, Hosseini H, Amin Ghobadi M, Afshani SM, Mirhassani R, Gohari K, Havasi F, Abdolghaffari A, Hedayatjoo B, Ghanei M. Immunogenicity and safety evaluation of a newly manufactured recombinant Baculovirus-Expressed quadrivalent influenza vaccine in adults 18 years old and Above: An Open-Label, phase III extension study. Int Immunopharmacol 2024; 136:112214. [PMID: 38823176 DOI: 10.1016/j.intimp.2024.112214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 06/03/2024]
Abstract
In the face of global health threats, there is a growing demand for vaccines that can be manufactured on a large scale within compressed timeline. This study responds to this imperative by delving into the evaluation of FluGuard, a novel recombinant influenza vaccine developed by Nivad Pharmed Salamat Company in Iran. Positioned as a phase 3 extension, the research aimed to evaluate the safety and immunogenicity of FluGuard in volunteers aged 18 and above. The study was conducted as a single-center, open-label clinical trial. All eligible volunteers received FluGuard (2021-2022 Formula) on day 0. Safety assessments occurred at days 1, 4, 7, 14, 28 and 42 post-vaccination. Immunogenicity was measured through seroconversion, seroprotection, and geometric mean titer fold increase in subgroups of 250 volunteers. Among the 4,260 volunteers were screened and assessed for eligibility, 1000 were enrolled. At day 28 post-vaccination, seroconversion rates for A/H1N1, A/H3N2, B/Yamagata, B/Victoria were 53.4 % [95 %CI: 46.7-60], 57.7 % [95 %CI: 51.1-64.3], 54.3 % [95 %CI: 47.7-60.9], and 36.2 % [95 %CI: 29.8-42.6], respectively in volunteers 18 years and above. The most common solicited adverse events were pain at the injection site, malaise, and headache. No suspected unexpected adverse events and adverse events of special interest occurred during the study period. Our findings suggested that FluGuard® exhibits a desirable safety profile and provides sufficient immunogenicity against influenza virus types A and B. However, extended studies are warranted to assess the long-term protective efficacy. Trial Registration: The study protocol was accepted by Iranian registry of clinical trial; https://www.irct.ir; IRCT20201104049265N2.
Collapse
Affiliation(s)
| | - Setayesh Sadeghi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Hosseini
- Clinical Trial Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Reihaneh Mirhassani
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran; Nivad Pharmed Salamat, Biotechnology Research Center, Tehran, Iran
| | - Kimiya Gohari
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Forugh Havasi
- Nivad Pharmed Salamat, Biotechnology Research Center, Tehran, Iran; Department of Chemistry, Faculty of Sciences, University of Kurdistan, Sanandaj, Iran
| | - Amirhossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Mostafa Ghanei
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Stein AN, Mills CW, McGovern I, McDermott KW, Dean A, Bogdanov AN, Sullivan SG, Haag MDM. Relative Vaccine Effectiveness of Cell- vs Egg-Based Quadrivalent Influenza Vaccine Against Test-Confirmed Influenza Over 3 Seasons Between 2017 and 2020 in the United States. Open Forum Infect Dis 2024; 11:ofae175. [PMID: 38698895 PMCID: PMC11064727 DOI: 10.1093/ofid/ofae175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
Background Influenza vaccine viruses grown in eggs may acquire egg-adaptive mutations that may reduce antigenic similarity between vaccine and circulating influenza viruses and decrease vaccine effectiveness. We compared cell- and egg-based quadrivalent influenza vaccines (QIVc and QIVe, respectively) for preventing test-confirmed influenza over 3 US influenza seasons (2017-2020). Methods Using a retrospective test-negative design, we estimated the relative vaccine effectiveness (rVE) of QIVc vs QIVe among individuals aged 4 to 64 years who had an acute respiratory or febrile illness and were tested for influenza in routine outpatient care. Exposure, outcome, and covariate data were obtained from electronic health records linked to pharmacy and medical claims. Season-specific rVE was estimated by comparing the odds of testing positive for influenza among QIVc vs QIVe recipients. Models were adjusted for age, sex, geographic region, influenza test date, and additional unbalanced covariates. A doubly robust approach was used combining inverse probability of treatment weights with multivariable regression. Results The study included 31 824, 33 388, and 34 398 patients in the 2017-2018, 2018-2019, and 2019-2020 seasons, respectively; ∼10% received QIVc and ∼90% received QIVe. QIVc demonstrated superior effectiveness vs QIVe in prevention of test-confirmed influenza: rVEs were 14.8% (95% CI, 7.0%-22.0%) in 2017-2018, 12.5% (95% CI, 4.7%-19.6%) in 2018-2019, and 10.0% (95% CI, 2.7%-16.7%) in 2019-2020. Conclusions This study demonstrated consistently superior effectiveness of QIVc vs QIVe in preventing test-confirmed influenza over 3 seasons characterized by different circulating viruses and degrees of egg adaptation.
Collapse
Affiliation(s)
- Alicia N Stein
- Centre for Outcomes Research and Epidemiology, CSL Seqirus, Melbourne, Australia
| | | | - Ian McGovern
- Centre for Outcomes Research and Epidemiology, CSL Seqirus, Waltham, Massachusetts, USA
| | | | - Alex Dean
- Real World Evidence, Veradigm, Chicago, Illinois, USA
| | | | - Sheena G Sullivan
- WHO Collaborating Centre for Reference and Research on Influenza, Royal Melbourne Hospital, and Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
- Department of Epidemiology, University of California, Los Angeles, California, USA
| | - Mendel D M Haag
- Centre for Outcomes Research and Epidemiology, CSL Seqirus, Amsterdam, Netherlands
| |
Collapse
|
10
|
Li H, Sun Z, Zheng T, Chen M, Lei X, Yu X, Ning Z. CD46 inhibits the replication of swine influenza viruses by promoting the production of type I IFNs in PK-15 cells. Vet Res Commun 2024; 48:1111-1119. [PMID: 38153594 DOI: 10.1007/s11259-023-10289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
Swine flu caused by swine influenza A virus (swIAV) is an acute respiratory viral disease that is spreading in swine herds worldwide. Although the effect of some host factors on replication of swIAV has been identified, the role of CD46 in this process is unclear. Here, we report that CD46 inhibits the replication of swIAV by promoting the production of type I interferons (IFNs) in porcine kidney (PK-15) cells. CD46 knockout (CD46-KO) and stably expressing (CD46-overexpression) PK-15 cells were prepared using lentivirus-mediated CRISPR/Cas9 gene editing and seamless cloning technology. The results of virus infection in CD46-overexpression PK-15 cells showed that the replication of H1N1 and H3N2 swIAVs were inhibited, and the production of type I IFNs (IFN-α, IFN-β), interferon regulatory factor (IRF) 3, and mitochondrial antiviral-signaling protein (MAVS) was enhanced. Virus infection in CD46-KO PK-15 cells showed the opposite results. Further results showed that CD46-KO PK-15 cells have a favorable ability to proliferate influenza viruses compared to Madin-Darby canine kidney (MDCK) and PK-15 cells. These findings indicate that CD46 acts as promising target regulating the replication of swIAV, and help to develop new agents against infection and replication of the virus.
Collapse
Affiliation(s)
- Huizi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenzhen Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Tingting Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ming Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoling Lei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xianglong Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhangyong Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
| |
Collapse
|
11
|
Zinnecker T, Badri N, Araujo D, Thiele K, Reichl U, Genzel Y. From single-cell cloning to high-yield influenza virus production - implementing advanced technologies in vaccine process development. Eng Life Sci 2024; 24:2300245. [PMID: 38584687 PMCID: PMC10991716 DOI: 10.1002/elsc.202300245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 04/09/2024] Open
Abstract
Innovations in viral vaccine manufacturing are crucial for pandemic preparedness and to meet ever-rising global demands. For influenza, however, production still mainly relies on technologies established decades ago. Although modern production shifts from egg-based towards cell culture technologies, the full potential has not yet been fully exploited. Here, we evaluate whether implementation of state-of-the-art technologies for cell culture-based recombinant protein production are capable to challenge outdated approaches in viral vaccine process development. For this, a fully automated single-cell cloning strategy was established to generate monoclonal suspension Madin-Darby canine kidney (MDCK) cells. Among selected cell clones, we could observe distinct metabolic and growth characteristics, with C59 reaching a maximum viable cell concentration of 17.3 × 106 cells/mL and low doubling times in batch mode. Screening for virus production using a panel of human vaccine-relevant influenza A and B viruses in an ambr15 system revealed high titers with yields competing or even outperforming available MDCK cell lines. With C113, we achieved cell-specific virus yields of up to 25,000 virions/cell, making this cell clone highly attractive for vaccine production. Finally, we confirmed process performance at a 50-fold higher working volume. In summary, we present a scalable and powerful approach for accelerated development of high-yield influenza virus production in chemically defined medium starting from a single cell.
Collapse
Affiliation(s)
- Tilia Zinnecker
- Max Planck Institute for Dynamics of Complex Technical SystemsMagdeburgGermany
| | | | - Diogo Araujo
- Sartorius Stedim Biotech S.A.Aubagne CedexFrance
| | | | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical SystemsMagdeburgGermany
- Bioprocess EngineeringOtto‐von‐Guericke UniversityMagdeburgGermany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical SystemsMagdeburgGermany
| |
Collapse
|
12
|
Chua SCJH, Cui J, Sachaphibulkij K, Tan ISL, Tan HQ, Lim HM, Engelberg D, Lim LHK. The ER-Golgi transport of influenza virus through NS1-Sec13 association during virus replication. Microbiol Spectr 2024; 12:e0260923. [PMID: 38038453 PMCID: PMC10782970 DOI: 10.1128/spectrum.02609-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
IMPORTANCE Influenza A virus is a respiratory virus that can cause complications such as acute bronchitis and secondary bacterial pneumonia. Drug therapies and vaccines are available against influenza, albeit limited by drug resistance and the non-universal vaccine administration. Hence there is a need for host-targeted therapies against influenza to provide an effective alternative therapeutic target. Sec13 was identified as a novel host interactor of influenza. Endoplasmic reticulum-to-Golgi transport is an important pathway of influenza virus replication and viral export. Specifically, Sec13 has a functional role in influenza replication and virulence.
Collapse
Affiliation(s)
- Sonja C. J. H. Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
- CREATE-NUS-HUJ Molecular Mechanisms of Inflammatory Diseases Programme, National University of Singapore, Singapore, Singapore
- Department of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jianzhou Cui
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Karishma Sachaphibulkij
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Isabelle Siang Ling Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Hui Qing Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Hong Meng Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - David Engelberg
- CREATE-NUS-HUJ Molecular Mechanisms of Inflammatory Diseases Programme, National University of Singapore, Singapore, Singapore
- Department of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lina H. K. Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore, Singapore
| |
Collapse
|
13
|
Liu F, Gross FL, Joshi S, Gaglani M, Naleway AL, Murthy K, Groom HC, Wesley MG, Edwards LJ, Grant L, Kim SS, Sambhara S, Gangappa S, Tumpey T, Thompson MG, Fry AM, Flannery B, Dawood FS, Levine MZ. Redirecting antibody responses from egg-adapted epitopes following repeat vaccination with recombinant or cell culture-based versus egg-based influenza vaccines. Nat Commun 2024; 15:254. [PMID: 38177116 PMCID: PMC10767121 DOI: 10.1038/s41467-023-44551-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Abstract
Repeat vaccination with egg-based influenza vaccines could preferentially boost antibodies targeting the egg-adapted epitopes and reduce immunogenicity to circulating viruses. In this randomized trial (Clinicaltrials.gov: NCT03722589), sera pre- and post-vaccination with quadrivalent inactivated egg-based (IIV4), cell culture-based (ccIIV4), and recombinant (RIV4) influenza vaccines were collected from healthcare personnel (18-64 years) in 2018-19 (N = 723) and 2019-20 (N = 684) influenza seasons. We performed an exploratory analysis. Vaccine egg-adapted changes had the most impact on A(H3N2) immunogenicity. In year 1, RIV4 induced higher neutralizing and total HA head binding antibodies to cell- A(H3N2) virus than ccIIV4 and IIV4. In year 2, among the 7 repeat vaccination arms (IIV4-IIV4, IIV4-ccIIV4, IIV4-RIV4, RIV4-ccIIV4, RIV4-RIV4, ccIIV4-ccIIV4 and ccIIV4-RIV4), repeat vaccination with either RIV4 or ccIIV4 further improved antibody responses to circulating viruses with decreased neutralizing antibody egg/cell ratio. RIV4 also had higher post-vaccination A(H1N1)pdm09 and A(H3N2) HA stalk antibodies in year 1, but there was no significant difference in HA stalk antibody fold rise among vaccine groups in either year 1 or year 2. Multiple seasons of non-egg-based vaccination may be needed to redirect antibody responses from immune memory to egg-adapted epitopes and re-focus the immune responses towards epitopes on the circulating viruses to improve vaccine effectiveness.
Collapse
Affiliation(s)
- Feng Liu
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - F Liaini Gross
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Sneha Joshi
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Manjusha Gaglani
- Baylor Scott & White Health, Temple, TX, USA
- Baylor College of Medicine, Temple, TX, USA
- Texas A & M University, College of Medicine, Temple, TX, USA
| | - Allison L Naleway
- Kaiser Permanente Northwest Center for Health Research, Portland, OR, USA
| | | | - Holly C Groom
- Kaiser Permanente Northwest Center for Health Research, Portland, OR, USA
| | - Meredith G Wesley
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
- Abt Associates, Atlanta, GA, USA
| | | | - Lauren Grant
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Sara S Kim
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Terrence Tumpey
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mark G Thompson
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Alicia M Fry
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Brendan Flannery
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Fatimah S Dawood
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Min Z Levine
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
14
|
Kitikoon P, Knetter SM, Mogler MA, Morgan CL, Hoehn A, Puttamreddy S, Strait EL, Segers RPAM. Quadrivalent neuraminidase RNA particle vaccine protects pigs against homologous and heterologous strains of swine influenza virus infection. Vaccine 2023; 41:6941-6951. [PMID: 37884412 DOI: 10.1016/j.vaccine.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 09/23/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023]
Abstract
Influenza A virus in swine (IAV-S) continues to cause significant negative impact to both sows and growing pigs. The viral hemagglutinin (HA) and neuraminidase (NA) genes continue to evolve with HA diversifying at a faster rate than NA. Depending on country, whole inactivated virus (WIV) commercial and autogenous vaccines, as well as veterinary prescription vaccines targeting HA, are currently available. The use of these vaccines is focused on reducing virus and clinical signs in sows and to provide HA-specific maternally derived antibodies (MDA) to their suckling pigs. The deficiency in this strategy is that HA-MDA does not persist long enough to protect pigs through their growing phase from infection, and HA-MDA can interfere with effective pig immunization. This study evaluated the immunogenicity and efficacy of an adjuvanted, quadrivalent RNA Particle vaccine (Sequivity NA), currently licensed as Sequivity® IAV-S NA. This vaccine was formulated based on four NA antigens representing the major NA clades of IAV subtypes H1N1, H1N2 and H3N2 circulating in swine herds in the United States. In a series of trials, pigs were vaccinated twice, at three days and three weeks of age (WOA), followed by challenge with either homologous or heterologous IAV strains at 8 or 15 WOA. The Sequivity NA vaccine induced robust serum NA inhibition (NI) antibody and protected against IAV-S strains with homologous and heterologous NA to that of the vaccine. The magnitude and duration of nasal shedding was reduced in vaccinated-pigs challenged with either homologous or heterologous virus within the same NA clade. This NA-based RNA Particle vaccine avoids the known impact of HA-MDA on pig vaccination and provides a new tool to successfully reduce IAV-induced disease in the pig population.
Collapse
Affiliation(s)
| | | | | | | | - Allison Hoehn
- Merck Animal Health, De Soto, KS 66018, United States
| | | | - Erin L Strait
- Merck Animal Health, De Soto, KS 66018, United States.
| | | |
Collapse
|
15
|
Coleman BL, Gutmanis I, McGovern I, Haag M. Effectiveness of Cell-Based Quadrivalent Seasonal Influenza Vaccine: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2023; 11:1607. [PMID: 37897009 PMCID: PMC10610589 DOI: 10.3390/vaccines11101607] [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: 08/24/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Cell-based seasonal influenza vaccine viruses may more closely match recommended vaccine strains than egg-based options. We sought to evaluate the effectiveness of seasonal cell-based quadrivalent influenza vaccine (QIVc), as reported in the published literature. A systematic literature review was conducted (PROSPERO CRD42020160851) to identify publications reporting on the effectiveness of QIVc in persons aged ≥6 months relative to no vaccination or to standard-dose, egg-based quadrivalent or trivalent influenza vaccines (QIVe/TIVe). Publications from between 1 January 2016 and 25 February 2022 were considered. The review identified 18 relevant publications spanning three influenza seasons from the 2017-2020 period, with an overall pooled relative vaccine effectiveness (rVE) of 8.4% (95% CI, 6.5-10.2%) for QIVc vs. QIVe/TIVe. Among persons aged 4-64 years, the pooled rVE was 16.2% (95% CI, 7.6-24.8%) for 2017-2018, 6.1% (4.9-7.3%) for 2018-2019, and 10.1% (6.3-14.0%) for 2019-2020. For adults aged ≥65 years, the pooled rVE was 9.9% (95% CI, 6.9-12.9%) in the egg-adapted 2017-2018 season, whereas there was no significant difference in 2018-2019. For persons aged 4-64 years, QIVc was consistently more effective than QIVe/TIVe over the three influenza seasons. For persons aged ≥65 years, protection with QIVc was greater than QIVe or TIVe during the 2017-2018 season and comparable in 2018-2019.
Collapse
Affiliation(s)
- Brenda L. Coleman
- Sinai Health, Toronto, ON M5G 2A2, Canada;
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Iris Gutmanis
- Sinai Health, Toronto, ON M5G 2A2, Canada;
- Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | | | - Mendel Haag
- CSL Seqirus, 1105 BJ Amsterdam, The Netherlands
| |
Collapse
|
16
|
Domnich A, Amicizia D, Lai PL, Ogliastro M, Piedrahita-Tovar M, Orsi A, Icardi G, Panatto D. Three seasons of enhanced safety surveillance of a cell culture-based quadrivalent influenza vaccine. Hum Vaccin Immunother 2023; 19:2261689. [PMID: 37787067 PMCID: PMC10549188 DOI: 10.1080/21645515.2023.2261689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
The objective of this paper is to summarize annual enhanced safety surveillance activity across three seasons (2019/20-2021/22) for cell culture-based quadrivalent influenza vaccine (QIVc; Flucelvax® Tetra) in all age groups. This activity was conducted in primary care setting in Genoa (Italy) during the seasons 2019/20, 2020/21 and 2021/22. All adverse events registered within the first seven days following immunization were analyzed by season, type, age group and seriousness. Over three seasons, 3,603 QIVc exposures were recorded within the enhanced passive safety surveillance activity. No safety signals were identified. The overall reporting rates of individual case safety reports for the seasons 2019/20, 2020/21 and 2021/22 were 1.75%, 0.48% and 0.40%, respectively. The average number of adverse events per individual case safety report was similar (range 3.3-3.8 adverse events per case report) across the three seasons. Most adverse events were reactogenic in nature. The rate of adverse events was similarly low in all age groups. Enhanced passive safety surveillance activity is a feasible approach for the post-marketing monitoring of seasonal influenza vaccines. Within its limitations, results of this study support the favorable safety profile of QIVc. These safety data could further bolster public trust in influenza vaccines with the goal to increase vaccination uptake in all target groups.
Collapse
Affiliation(s)
- Alexander Domnich
- Hygiene Unit, San Martino Polyclinic Hospital-IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Daniela Amicizia
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Interuniversity Research Center on Influenza and Other Transmissible Infections (CIRI-IT), Genoa, Italy
| | - Piero Luigi Lai
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Interuniversity Research Center on Influenza and Other Transmissible Infections (CIRI-IT), Genoa, Italy
| | - Matilde Ogliastro
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | | | - Andrea Orsi
- Hygiene Unit, San Martino Polyclinic Hospital-IRCCS for Oncology and Neurosciences, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Interuniversity Research Center on Influenza and Other Transmissible Infections (CIRI-IT), Genoa, Italy
| | - Giancarlo Icardi
- Hygiene Unit, San Martino Polyclinic Hospital-IRCCS for Oncology and Neurosciences, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Interuniversity Research Center on Influenza and Other Transmissible Infections (CIRI-IT), Genoa, Italy
| | - Donatella Panatto
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
- Interuniversity Research Center on Influenza and Other Transmissible Infections (CIRI-IT), Genoa, Italy
| |
Collapse
|
17
|
Imran M, Puig-Barbera J, Ortiz JR, Lopez-Gonzalez L, Dean A, Bonafede M, Haag M. Relative Effectiveness of the Cell-Based Quadrivalent Influenza Vaccine in Preventing Cardiorespiratory Hospitalizations in Adults Aged 18-64 Years During the 2019-2020 US Influenza Season. Open Forum Infect Dis 2023; 10:ofad304. [PMID: 37496602 PMCID: PMC10368203 DOI: 10.1093/ofid/ofad304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/05/2023] [Indexed: 07/28/2023] Open
Abstract
Background The mammalian cell-based quadrivalent inactivated influenza vaccine (IIV4c) has advantages over egg-based quadrivalent inactivated influenza vaccine (IIV4e), as production using cell-derived candidate viruses eliminates the opportunity for egg adaptation. This study estimated the relative vaccine effectiveness (rVE) of IIV4c versus IIV4e in preventing cardiorespiratory hospitalizations during the 2019-2020 US influenza season. Methods We conducted a retrospective cohort study using electronic medical records linked to claims data of US individuals aged 18-64 years. We assessed rVE against cardiorespiratory hospitalizations and against subcategories of this outcome, including influenza, pneumonia, myocardial infarction and ischemic stroke, and respiratory hospitalizations. We used a doubly robust inverse probability of treatment weighting and logistic regression model to obtain odds ratios (ORs; odds of outcome among IIV4c recipients/odds of outcome among IIV4e recipients) adjusted for age, sex, race, ethnicity, geographic region, vaccination week, health status, frailty, and healthcare resource utilization. rVE was calculated as 100(1 - ORadjusted). Results In total, 1 491 097 individuals (25.2%) received IIV4c, and 4 414 758 (74.8%) received IIV4e. IIV4c was associated with lower odds of cardiorespiratory (rVE, 2.5% [95% confidence interval, 0.9%-4.1%]), respiratory (3.7% [1.5%-5.8%]), and influenza (9.3% [0.4%-17.3%]) hospitalizations among adults 18-64 years of age. No difference was observed for the other outcomes. Conclusions This real-world study conducted for the 2019-2020 season demonstrated that vaccination with IIV4c was associated with fewer cardiorespiratory, respiratory, and influenza hospitalizations compared with IIV4e.
Collapse
Affiliation(s)
- Mahrukh Imran
- Correspondence: Mahrukh Imran, MScPH, CSL Seqirus, Ste 504, 16766 TransCanada Hwy, Kirkland, QC H9H 4M7, Canada ()
| | | | - Justin R Ortiz
- University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | |
Collapse
|
18
|
Cadar AN, Martin DE, Bartley JM. Targeting the hallmarks of aging to improve influenza vaccine responses in older adults. Immun Ageing 2023; 20:23. [PMID: 37198683 PMCID: PMC10189223 DOI: 10.1186/s12979-023-00348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Abstract
Age-related declines in immune response pose a challenge in combating diseases later in life. Influenza (flu) infection remains a significant burden on older populations and often results in catastrophic disability in those who survive infection. Despite having vaccines designed specifically for older adults, the burden of flu remains high and overall flu vaccine efficacy remains inadequate in this population. Recent geroscience research has highlighted the utility in targeting biological aging to improve multiple age-related declines. Indeed, the response to vaccination is highly coordinated, and diminished responses in older adults are likely not due to a singular deficit, but rather a multitude of age-related declines. In this review we highlight deficits in the aged vaccine responses and potential geroscience guided approaches to overcome these deficits. More specifically, we propose that alternative vaccine platforms and interventions that target the hallmarks of aging, including inflammation, cellular senescence, microbiome disturbances, and mitochondrial dysfunction, may improve vaccine responses and overall immunological resilience in older adults. Elucidating novel interventions and approaches that enhance immunological protection from vaccination is crucial to minimize the disproportionate effect of flu and other infectious diseases on older adults.
Collapse
Affiliation(s)
- Andreia N Cadar
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Dominique E Martin
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Jenna M Bartley
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
| |
Collapse
|
19
|
Peletta A, Lemoine C, Courant T, Collin N, Borchard G. Meeting vaccine formulation challenges in an emergency setting: Towards the development of accessible vaccines. Pharmacol Res 2023; 189:106699. [PMID: 36796463 DOI: 10.1016/j.phrs.2023.106699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Vaccination is considered one of the most successful strategies to prevent infectious diseases. In the event of a pandemic or epidemic, the rapid development and distribution of the vaccine to the population is essential to reduce mortality, morbidity and transmission. As seen during the COVID-19 pandemic, the production and distribution of vaccines has been challenging, in particular for resource-constrained settings, essentially slowing down the process of achieving global coverage. Pricing, storage, transportation and delivery requirements of several vaccines developed in high-income countries resulted in limited access for low-and-middle income countries (LMICs). The capacity to manufacture vaccines locally would greatly improve global vaccine access. In particular, for the development of classical subunit vaccines, the access to vaccine adjuvants is a pre-requisite for more equitable access to vaccines. Vaccine adjuvants are agents required to augment or potentiate, and possibly target the specific immune response to such type of vaccine antigens. Openly accessible or locally produced vaccine adjuvants may allow for faster immunization of the global population. For local research and development of adjuvanted vaccines to expand, knowledge on vaccine formulation is of paramount importance. In this review, we aim to discuss the optimal characteristics of a vaccine developed in an emergency setting by focusing on the importance of vaccine formulation, appropriate use of adjuvants and how this may help overcome barriers for vaccine development and production in LMICs, achieve improved vaccine regimens, delivery and storage requirements.
Collapse
Affiliation(s)
- Allegra Peletta
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| | - Céline Lemoine
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Thomas Courant
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Institute, Rue du Champ-Blanchod 4, 1228 Plan-les-Ouates, Switzerland.
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, Rue Michel-Servet 1, 1221 Geneva, Switzerland.
| |
Collapse
|
20
|
Nucleoside-Modified mRNA-Based Influenza Vaccines Circumvent Problems Associated with H3N2 Vaccine Strain Egg Adaptation. J Virol 2023; 97:e0172322. [PMID: 36533954 PMCID: PMC9888232 DOI: 10.1128/jvi.01723-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Most human influenza vaccine antigens are produced in fertilized chicken eggs. Recent H3N2 egg-based vaccine antigens have limited effectiveness, partially due to egg-adaptive substitutions that alter the antigenicity of the hemagglutinin (HA) protein. The nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP) vaccine platform is a promising alternative for egg-based influenza vaccines because mRNA-LNP-derived antigens are not subject to adaptive pressures that arise during the production of antigens in chicken eggs. Here, we compared H3N2-specific antibody responses in mice vaccinated with either 3c.2A H3-encoding mRNA-LNP or a conventional egg-based Fluzone vaccine (which included an egg-adapted 3c.2A antigen) supplemented with an MF59-like adjuvant. We tested mRNA-LNP encoding wild-type and egg-adapted H3 antigens. We found that mRNA-LNP encoding wild-type H3 elicited antibodies that neutralized the wild-type 3c.2A H3N2 virus more effectively than antibodies elicited by mRNA-LNP encoding egg-adapted H3 or the egg-based Fluzone vaccine. mRNA-LNP expressing either wild-type or egg-adapted H3 protected mice against infection with the wild-type 3c2.A H3N2, whereas the egg-based Fluzone vaccine did not. We found that both mRNA-LNP vaccines elicited high levels of group 2 HA stalk-reactive antibodies, which likely contributed to protection in vivo. Our studies indicate that nucleoside-modified mRNA-LNP-based vaccines can circumvent problems associated with egg adaptations with recent 3c2.A H3N2 viruses. IMPORTANCE This study shows that the nucleoside-modified mRNA-LNP vaccine platform is a promising alternative for egg-based influenza vaccines. We show that mRNA-LNP vaccines expressing H3 antigens elicit high levels of antibodies in mice and protect against H3N2 influenza virus infection.
Collapse
|
21
|
Rockman S, Laurie K, Ong C, Rajaram S, McGovern I, Tran V, Youhanna J. Cell-Based Manufacturing Technology Increases Antigenic Match of Influenza Vaccine and Results in Improved Effectiveness. Vaccines (Basel) 2022; 11:52. [PMID: 36679895 PMCID: PMC9861528 DOI: 10.3390/vaccines11010052] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
To ensure that vaccination offers the best protection against an infectious disease, sequence identity between the vaccine and the circulating strain is paramount. During replication of nucleic acid, random mutations occur due to the level of polymerase fidelity. In traditional influenza vaccine manufacture, vaccine viruses are propagated in fertilized chicken eggs, which can result in egg-adaptive mutations in the antigen-encoding genes. Whilst this improves infection and replication in eggs, mutations may reduce the effectiveness of egg-based influenza vaccines against circulating human viruses. In contrast, egg-adaptive mutations are avoided when vaccine viruses are propagated in Madin-Darby canine kidney (MDCK) cell lines during manufacture of cell-based inactivated influenza vaccines. The first mammalian cell-only strain was included in Flucelvax® Quadrivalent in 2017. A sequence analysis of the viruses selected for inclusion in this vaccine (n = 15 vaccine strains, containing both hemagglutinin and neuraminidase) demonstrated that no mutations occur in the antigenic sites of either hemagglutinin or neuraminidase, indicating that cell adaptation does not occur during production of this cell-based vaccine. The development of this now entirely mammalian-based vaccine system, which incorporates both hemagglutinin and neuraminidase, ensures that the significant protective antigens are equivalent to the strains recommended by the World Health Organization (WHO) in both amino acid sequence and glycosylation pattern. The inclusion of both proteins in a vaccine may provide an advantage over recombinant vaccines containing hemagglutinin alone. Findings from real world effectiveness studies support the use of cell-based influenza vaccines.
Collapse
Affiliation(s)
- Steven Rockman
- CSL Seqirus Ltd., Parkville, VIC 3050, Australia
- Department of Immunology and Microbiology, The University of Melbourne, Parkville, VIC 3050, Australia
| | - Karen Laurie
- CSL Seqirus Ltd., Parkville, VIC 3050, Australia
| | - Chi Ong
- CSL Seqirus Ltd., Parkville, VIC 3050, Australia
| | | | | | - Vy Tran
- CSL Seqirus Ltd., Kirkland, QC H9H 4M7, Canada
| | | |
Collapse
|
22
|
Robinson C, Van Boxmeer J, Tilson H, Scialli A, Vanchiere JA, Ides E, Sawlwin D, Molrine D, Hohenboken M, Edelman J, Albano JD. Outcomes in Pregnant Persons Immunized with a Cell-Based Quadrivalent Inactivated Influenza Vaccine: A Prospective Observational Cohort Study. Vaccines (Basel) 2022; 10:1600. [PMID: 36298465 PMCID: PMC9612226 DOI: 10.3390/vaccines10101600] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Objective: To evaluate pregnancy and infant outcomes among persons immunized with a cell-based quadrivalent inactivated influenza vaccine (IIV4c) during routine pregnancy care. Design: Prospective observational cohort. Setting: US-based obstetrics/gynecology clinics. Population: Pregnant persons. This US-based, prospective observational cohort study evaluated the safety of quadrivalent inactivated influenza vaccine (IIV4c; Flucelvax® Quad) in pregnant persons immunized over 3 influenza seasons between 2017 and 2020. Pregnant persons were immunized with IIV4c as part of routine care, after which their health care provides HCPs with all observational data to a single coordinating center. Follow-up data were collected at the end of the second trimester and/or at the time of pregnancy outcome. A scientific advisory committee reviewed the data. Prevalence point estimates were reported with 95% confidence intervals (CIs). Pregnancy outcomes included: live birth, stillbirth, spontaneous abortion, elective termination, and maternal death. Infant outcomes included: preterm birth (<37 weeks gestational age), low birth weight (<2500 g), or major congenital malformations (MCMs). Of the 665 evaluable participants, 659 (99.1%) had a live birth. No stillbirths (0% [95% CI 0.0−0.6]), 4 spontaneous abortions (1.9% [0.5−4.8]), and 1 elective termination (0.5% [0.0−2.6]) were reported. Among 673 infants, 9.2% (upper 95% CI 11.5%) were born prematurely, 5.8% (upper 95% CI 7.6%) had low birth weight, and 1.9% (upper 95% CI 3.1%) were reported to have an MCM. No maternal deaths were reported. Of the 2 infants who died shortly after birth, one was adjudicated as not related to the vaccine; the other’s cause could not be determined due to maternal loss to follow-up. The prevalence of adverse pregnancy outcomes or preterm birth, low birth weight, or MCMs in newborns was similar in persons vaccinated with IIV4c compared to the rates observed in US surveillance systems. The safety profile of IIV4c in pregnant persons is consistent with previously studied influenza vaccines.
Collapse
Affiliation(s)
| | | | - Hugh Tilson
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - John A. Vanchiere
- Louisiana State University Health Science Center, Shreveport, LA 71106, USA
| | - Ellis Ides
- Seqirus Netherlands B.V., 1105 BJ Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
23
|
Metabolic reprogramming and alteration of the redox state in hyper-productive MDCK cells for influenza a virus production. Biologicals 2022; 80:35-42. [PMID: 36114098 DOI: 10.1016/j.biologicals.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Influenza is a global public health issue leading to widespread morbidity and mortality with devastating economic loss annually. Madin-Darby Canine Kidney (MDCK) cell line has been a major cell line for influenza vaccine applications. Though many details of the host metabolic responses upon influenza A virus (IAV) infection have been documented, little is known about the metabolic reprogramming features of a hyper-productive host for IAV vaccine production. In this study, a MDCK cell clone H1 was shown to have a particular high productivity of 30 × 103 virions/cell. The glucose and amino acid metabolism of H1 were evaluated, indicating that the high producer had a particular metabolic reprogramming phenotype compared to its parental cell line (P): elevated glucose uptake, superior tricarboxylic acid cycle flux, moderate amino acid consumption, and better regulation of reactive oxygen species. Combined with the stronger mitochondrial function and mild antiviral and inflammatory responses characterized previously, our results indicated that the high producer had a sufficient intracellular energy supply, and balanced substrate distribution for IAV and host protein synthesis as well as the intracellular redox status. Understanding of these metabolic alterations paves the way for the rational cell line development and reasonable process optimization for high-yield influenza vaccine production.
Collapse
|
24
|
Gil A, Sinilaite A, Papenburg J. Summary of the National Advisory Committee on Immunization (NACI) Supplemental Statement on Recombinant Influenza Vaccines. CANADA COMMUNICABLE DISEASE REPORT = RELEVE DES MALADIES TRANSMISSIBLES AU CANADA 2022; 48:383-391. [PMID: 38116047 PMCID: PMC10729781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Background Recombinant protein technology is a novel platform for influenza vaccine manufacturing that differs significantly from existing egg-based and mammalian cell culture-based technologies. Supemtek™ is the first and, to date, the only recombinant quadrivalent influenza vaccine (RIV4) authorized for use in Canada in adults aged 18 years and older. The objective is to review the available evidence for efficacy, effectiveness, immunogenicity and safety of RIV4, and to summarize the National Advisory Committee on Immunization (NACI) recommendation regarding the use of Supemtek. Methods A systematic literature review and meta-analysis on the vaccine efficacy, effectiveness, immunogenicity and safety of RIV4 in adults was conducted according to methodology specified a priori in a written protocol. NACI evidence-based process was used to assess the available evidence and develop a recommendation regarding the use of Supemtek. Results Ten eligible studies were included in the evidence synthesis. One randomized controlled trial (RCT) in adults aged 50 years and older provided evidence that RIV4 may potentially offer improved protection against laboratory-confirmed influenza A infection compared to standard egg-based influenza vaccines. Data from eight RCTs assessing immunogenicity and five RCTs and one post-marketing surveillance study assessing safety indicated that Supemtek is a safe, well tolerated, and immunogenic alternative to conventional egg-based influenza vaccines for adults. Conclusion There is fair evidence that Supemtek is effective, safe, and has non-inferior immunogenicity to comparable vaccines, based on direct evidence in adults 18 years of age and older; thus, NACI recommends that Supemtek may be considered among the seasonal influenza vaccines offered to adults 18 years of age and older for their annual influenza vaccination.
Collapse
Affiliation(s)
- Anabel Gil
- Centre for Immunization Readiness, Public Health Agency of Canada, Ottawa, ON
| | - Angela Sinilaite
- Centre for Immunization Readiness, Public Health Agency of Canada, Ottawa, ON
| | - Jesse Papenburg
- NACI Influenza Working Group Chair
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Montréal Children's Hospital of the McGill University Health Centre, Montréal, QC
- Division of Microbiology, Department of Clinical Laboratory Medicine, Optilab Montréal - McGill University Health Centre, Montréal, QC
- Department of Epidemiology, Biostatistics, and Occupational Health, School of Population and Global Health, McGill University, Montréal, QC
| |
Collapse
|
25
|
Imran M, Ortiz JR, McLean HQ, Fisher L, O’Brien D, Bonafede M, Mansi JA, Boikos C. Relative Effectiveness of Cell-based Versus Egg-based Quadrivalent Influenza Vaccines in Children and Adolescents in the United States During the 2019-2020 Influenza Season. Pediatr Infect Dis J 2022; 41:769-774. [PMID: 35797705 PMCID: PMC9359763 DOI: 10.1097/inf.0000000000003620] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Egg-based influenza vaccine production can lead to the accumulation of mutations that affect antigenicity. The mammalian cell-based inactivated quadrivalent influenza vaccine (IIV4c) may improve effectiveness compared with egg-based vaccines. This study estimated the relative vaccine effectiveness (rVE) of IIV4c versus egg-based inactivated quadrivalent influenza vaccine (IIV4e) in preventing influenza-related medical encounters (IRME) among children and adolescents during the 2019-2020 US influenza season. METHODS This retrospective cohort study used a dataset linking primary and specialty care electronic medical records with medical and pharmacy claims data from US residents 4 through 17 years of age vaccinated with IIV4c or IIV4e during the 2019-2020 influenza season. Odds ratios (ORs) were derived from a doubly robust inverse probability of treatment-weighted approach adjusting for age, sex, race, ethnicity, region, index week, health status and two proxy variables for healthcare accessibility and use. Adjusted rVE was estimated by (1-OR adjusted )*100, and an exploratory analysis evaluated IRMEs separately for outpatient and inpatient settings. RESULTS The final study cohort included 60,480 (IIV4c) and 1,240,990 (IIV4e) vaccine recipients. Fewer IRMEs were reported in subjects vaccinated with IIV4c than IIV4e. The rVE for IIV4c versus IIV4e was 12.2% [95% confidence interval (CI): 7.5-16.6] for any IRME and 14.3% (9.3-19.0) for outpatient IRMEs. Inpatient IRMEs were much less frequent, and effectiveness estimates were around the null. CONCLUSIONS Fewer IRMEs occurred in pediatric subjects vaccinated with IIV4c versus IIV4e. These results support the greater effectiveness of IIV4c over IIV4e in this population during the 2019-2020 US influenza season.
Collapse
Affiliation(s)
| | - Justin R. Ortiz
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Huong Q McLean
- Marshfield Clinic Research Institute, Marshfield, Wisconsin
| | | | | | | | - James A. Mansi
- Seqirus Inc., Kirkland, Quebec, Canada, During Study Conduct
| | | |
Collapse
|
26
|
Chia MY, Lin CY, Chen PL, Lai CC, Weng TC, Sung WC, Hu AYC, Lee MS. Characterization and Immunogenicity of Influenza H7N9 Vaccine Antigens Produced Using a Serum-Free Suspension MDCK Cell-Based Platform. Viruses 2022; 14:v14091937. [PMID: 36146744 PMCID: PMC9502495 DOI: 10.3390/v14091937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Human infections with avian-origin H7N9 influenza A viruses were first reported in China, and an approximately 38% human mortality rate was described across six waves from February 2013 to September 2018. Vaccination is one of the most cost-effective ways to reduce morbidity and mortality during influenza epidemics and pandemics. Egg-based platforms for the production of influenza vaccines are labor-intensive and unable to meet the surging demand during pandemics. Therefore, cell culture-based technology is becoming the alternative strategy for producing influenza vaccines. The current influenza H7N9 vaccine virus (NIBRG-268), a reassortant virus from A/Anhui/1/2013 (H7N9) and egg-adapted A/PR/8/34 (H1N1) viruses, could grow efficiently in embryonated eggs but not mammalian cells. Moreover, a freezing-dry formulation of influenza H7N9 vaccines with long-term stability will be desirable for pandemic preparedness, as the occurrence of influenza H7N9 pandemics is not predictable. In this study, we adapted a serum-free anchorage-independent suspension Madin-Darby Canine Kidney (MDCK) cell line for producing influenza H7N9 vaccines and compared the biochemical characteristics and immunogenicity of three influenza H7N9 vaccine antigens produced using the suspension MDCK cell-based platform without freeze-drying (S-WO-H7N9), the suspension MDCK cell-based platform with freeze-drying (S-W-H7N9) or the egg-based platform with freeze-drying (E-W-H7N9). We demonstrated these three vaccine antigens have comparable biochemical characteristics. In addition, these three vaccine antigens induced robust and comparable neutralizing antibody (NT; geometric mean between 1016 and 4064) and hemagglutinin-inhibition antibody (HI; geometric mean between 640 and 1613) titers in mice. In conclusion, the serum-free suspension MDCK cell-derived freeze-dried influenza H7N9 vaccine is highly immunogenic in mice, and clinical development is warranted.
Collapse
Affiliation(s)
- Min-Yuan Chia
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Chun-Yang Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Po-Ling Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Chia-Chun Lai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Tsai-Chuan Weng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Wang-Chou Sung
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Alan Yung-Chih Hu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Min-Shi Lee
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
- Correspondence: ; Tel.: +886-(37)-246-166 (ext. 35520); Fax: +886-(37)-583-009
| |
Collapse
|
27
|
Pekarek MJ, Petro-Turnquist EM, Rubrum A, Webby RJ, Weaver EA. Expanding Mouse-Adapted Yamagata-like Influenza B Viruses in Eggs Enhances In Vivo Lethality in BALB/c Mice. Viruses 2022; 14:v14061299. [PMID: 35746770 PMCID: PMC9229684 DOI: 10.3390/v14061299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/10/2022] [Indexed: 01/23/2023] Open
Abstract
Despite the yearly global impact of influenza B viruses (IBVs), limited host range has been a hurdle to developing a readily accessible small animal disease model for vaccine studies. Mouse-adapting IBV can produce highly pathogenic viruses through serial lung passaging in mice. Previous studies have highlighted amino acid changes throughout the viral genome correlating with increased pathogenicity, but no consensus mutations have been determined. We aimed to show that growth system can play a role in mouse-adapted IBV lethality. Two Yamagata-lineage IBVs were serially passaged 10 times in mouse lungs before expansion in embryonated eggs or Madin-Darby canine kidney cells (London line) for use in challenge studies. We observed that virus grown in embryonated eggs was significantly more lethal in mice than the same virus grown in cell culture. Ten additional serial lung passages of one strain again showed virus grown in eggs was more lethal than virus grown in cells. Additionally, no mutations in the surface glycoprotein amino acid sequences correlated to differences in lethality. Our results suggest growth system can influence lethality of mouse-adapted IBVs after serial lung passaging. Further research can highlight improved mechanisms for developing animal disease models for IBV vaccine research.
Collapse
Affiliation(s)
- Matthew J. Pekarek
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.J.P.); (E.M.P.-T.)
| | - Erika M. Petro-Turnquist
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.J.P.); (E.M.P.-T.)
| | - Adam Rubrum
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.R.); (R.J.W.)
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (A.R.); (R.J.W.)
| | - Eric A. Weaver
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.J.P.); (E.M.P.-T.)
- Correspondence:
| |
Collapse
|
28
|
MADE: A Computational Tool for Predicting Vaccine Effectiveness for the Influenza A(H3N2) Virus Adapted to Embryonated Eggs. Vaccines (Basel) 2022; 10:vaccines10060907. [PMID: 35746515 PMCID: PMC9227319 DOI: 10.3390/vaccines10060907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 01/29/2023] Open
Abstract
Seasonal Influenza H3N2 virus poses a great threat to public health, but its vaccine efficacy remains suboptimal. One critical step in influenza vaccine production is the viral passage in embryonated eggs. Recently, the strength of egg passage adaptation was found to be rapidly increasing with time driven by convergent evolution at a set of functionally important codons in the hemagglutinin (HA1). In this study, we aim to take advantage of the negative correlation between egg passage adaptation and vaccine effectiveness (VE) and develop a computational tool for selecting the best candidate vaccine virus (CVV) for vaccine production. Using a probabilistic approach known as mutational mapping, we characterized the pattern of sequence evolution driven by egg passage adaptation and developed a new metric known as the adaptive distance (AD) which measures the overall strength of egg passage adaptation. We found that AD is negatively correlated with the influenza H3N2 vaccine effectiveness (VE) and ~75% of the variability in VE can be explained by AD. Based on these findings, we developed a computational package that can Measure the Adaptive Distance and predict vaccine Effectiveness (MADE). MADE provides a powerful tool for the community to calibrate the effect of egg passage adaptation and select more reliable strains with minimum egg-passaged changes as the seasonal A/H3N2 influenza vaccine.
Collapse
|
29
|
Hamamoto I, Takahashi H, Shimazaki N, Nakamura K, Mizuta K, Sato K, Nishimura H, Yamamoto N, Hasegawa H, Odagiri T, Tashiro M, Nobusawa E. Suitability of NIID-MDCK cells as a substrate for cell-based influenza vaccine development from the perspective of adventitious virus susceptibility. Microbiol Immunol 2022; 66:361-370. [PMID: 35545856 DOI: 10.1111/1348-0421.12985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/27/2022] [Accepted: 05/08/2022] [Indexed: 11/30/2022]
Abstract
The practical use of cell-based seasonal influenza vaccines is currently being considered in Japan. From the perspective of adventitious virus contamination, we assessed the suitability of NIID-MDCK cells (NIID-MDCK-Cs) as a safe substrate for the isolation of influenza viruses from clinical specimens. We first established a sensitive multiplex real-time PCR system to screen for 27 respiratory viruses and used it on 34 virus samples that were isolated by passaging influenza-positive clinical specimens in NIID-MDCK-Cs. Incidentally, the limit of detection of the system was 100 or fewer genome copies per reaction. In addition to influenza viruses, human enterovirus 68 (HEV-D68) genomes were detected in two samples after two or three passages in NIID-MDCK-Cs. To further investigate the susceptibility of NIID-MDCK-Cs to adventitious viruses, eight common respiratory viruses were subjected to passages in NIID-MDCK-Cs. The genome copy numbers of seven viruses other than parainfluenza 3 decreased below the limit of detection (LOD) by passage 4. By passaging in NIID-MDCK-Cs, the genome numbers of the input HEV-D68, 1 x 108 copies, declined to 102 at passage 3 and to under the LOD at passage 4, whereas those of the other six viruses were under the LOD by passage 3. These results implied that during the process of isolating influenza viruses with NIID-MDCK-Cs, contaminating viruses other than parainfluenza 3 can be efficiently removed by passages in NIID-MDCK-Cs. NIID-MDCK-Cs could be a safe substrate for isolating influenza viruses that can be used to develop cell-based influenza vaccine candidate viruses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Itsuki Hamamoto
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Hitoshi Takahashi
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Noriko Shimazaki
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Kazuya Nakamura
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Katsumi Mizuta
- Yamagata Prefectural Institute of Public Health, Yamagata, Japan
| | - Ko Sato
- Virus Research Center, Sendai Medical Center, National Hospital Organization, Sendai, Japan
| | - Hidekazu Nishimura
- Virus Research Center, Sendai Medical Center, National Hospital Organization, Sendai, Japan
| | - Norio Yamamoto
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan.,Department of Microbiology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Hideki Hasegawa
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Takato Odagiri
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Masato Tashiro
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| | - Eri Nobusawa
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayama-shi, Tokyo, 208-0011, Japan
| |
Collapse
|
30
|
Williams KV, Zhai B, Alcorn JF, Patricia Nowalk M, Levine MZ, Kim SS, Flannery B, Moehling Geffel K, Jaber Merranko A, Nagg JP, Collins M, Susick M, Clarke KS, Zimmerman RK, Martin JM. A randomized controlled trial of antibody response to 2019-20 cell-based inactivated and egg-based live attenuated influenza vaccines in children and young adults. Vaccine 2022; 40:780-788. [PMID: 34952751 PMCID: PMC8803136 DOI: 10.1016/j.vaccine.2021.12.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Hemagglutination inhibition (HAI) titers to the live-attenuated influenza vaccine (LAIV4) are typically lower than its counterpart egg-based inactivated influenza vaccines (IIV). Similar comparisons have not been made between LAIV4 and the 4-strain, cell-culture inactivated influenza vaccine (ccIIV4). We compared healthy children's and young adults' HAI titers against the 2019-2020 LAIV4 and ccIIV4. METHODS Participants aged 4-21 years were randomized 1:1 to receive ccIIV4 (n = 100) or LAIV4 (n = 98). Blood was drawn prevaccination and on day 28 (21-35) post vaccination. HAI assays against egg-grown A/H1N1, A/H3N2, both vaccine B strains and cell-grown A/H3N2 antigens were conducted. Primary outcomes were geometric mean titers (GMT) and geometric mean fold rise (GMFR) in titers. RESULTS GMTs to A/H1N1, A/H3N2 and B/Victoria increased following both ccIIV and LAIV and to B/Yamagata following ccIIV (p < 0.05). The GMFR range was 2.4-3.0 times higher for ccIIV4 than for LAIV4 (p < 0.001). Within vaccine types, egg-grown A/H3N2 GMTs were higher (p < 0.05) than cell-grown GMTs [ccIIV4 day 28: egg = 205 (95% CI: 178-237); cell = 136 (95% CI:113-165); LAIV4 day 28: egg = 96 (95% CI: 83-112); cell = 63 (95% CI: 58-74)]. The GMFR to A/H3N2 cell-grown and egg-grown antigens were similar. Pre-vaccination titers inversely predicted GMFR. CONCLUSION The HAI response to ccIIV4 was greater than LAIV4 in this study of mostly older children, and day 0 HAI titers inversely predicted GMFR for both vaccines. Lower prevaccination titers were associated with greater GMFR in both vaccine groups.
Collapse
Affiliation(s)
- Katherine V Williams
- Department of Family Medicine, University of Pittsburgh, 4420 Bayard Street, Suite 520, Pittsburgh, PA 15260, USA.
| | - Bo Zhai
- Department of Immunology, University of Pittsburgh, 9127 Rangos Research Center, 4401 Penn Avenue, Pittsburgh, PA 15224 USA.
| | - John F Alcorn
- Department of Immunology, University of Pittsburgh, 9127 Rangos Research Center, 4401 Penn Avenue, Pittsburgh, PA 15224 USA; Department of Pediatrics, University of Pittsburgh, 3520 Fifth Avenue, Pittsburgh, PA 15213, USA.
| | - Mary Patricia Nowalk
- Department of Family Medicine, University of Pittsburgh, 4420 Bayard Street, Suite 520, Pittsburgh, PA 15260, USA.
| | - Min Z Levine
- National Center Immunizations and Respiratory Disease, Center for Disease Control and Prevention, Atlanta, GA, USA.
| | - Sara S Kim
- National Center Immunizations and Respiratory Disease, Center for Disease Control and Prevention, Atlanta, GA, USA.
| | - Brendan Flannery
- National Center Immunizations and Respiratory Disease, Center for Disease Control and Prevention, Atlanta, GA, USA.
| | - Krissy Moehling Geffel
- Department of Family Medicine, University of Pittsburgh, 4420 Bayard Street, Suite 520, Pittsburgh, PA 15260, USA.
| | - Amanda Jaber Merranko
- Falk Pharmacy, University of Pittsburgh Medical Center (UPMC), 3601 Fifth Avenue, Pittsburgh, PA 15213, USA.
| | - Jennifer P Nagg
- Department of Pediatrics, University of Pittsburgh, 3520 Fifth Avenue, Pittsburgh, PA 15213, USA.
| | - Mark Collins
- Department of Family Medicine, University of Pittsburgh, 4420 Bayard Street, Suite 520, Pittsburgh, PA 15260, USA.
| | - Michael Susick
- Department of Family Medicine, University of Pittsburgh, 4420 Bayard Street, Suite 520, Pittsburgh, PA 15260, USA.
| | - Karen S Clarke
- Department of Family Medicine, University of Pittsburgh, 4420 Bayard Street, Suite 520, Pittsburgh, PA 15260, USA.
| | - Richard K Zimmerman
- Department of Family Medicine, University of Pittsburgh, 4420 Bayard Street, Suite 520, Pittsburgh, PA 15260, USA.
| | - Judith M Martin
- Department of Pediatrics, University of Pittsburgh, 3520 Fifth Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
31
|
Relative Effectiveness of Cell-Cultured versus Egg-Based Seasonal Influenza Vaccines in Preventing Influenza-Related Outcomes in Subjects 18 Years Old or Older: A Systematic Review and Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19020818. [PMID: 35055642 PMCID: PMC8775496 DOI: 10.3390/ijerph19020818] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 02/06/2023]
Abstract
Avian mutations in vaccine strains obtained from embryonated eggs could impair vaccine effectiveness. We performed a systematic review and meta-analysis of the adjusted relative vaccine effectiveness (arVE) of seed cell-cultured influenza vaccines (ccIV) compared to egg-based influenza vaccines (eIV) in preventing laboratory-confirmed influenza related outcomes (IRO) or IRO by clinical codes, in subjects 18 and over. We completed the literature search in January 2021; applied exclusion criteria, evaluated risk of bias of the evidence, and performed heterogeneity, publication bias, qualitative, quantitative and sensitivity analyses. All estimates were computed using a random approach. International Prospective Register of Systematic Reviews, CRD42021228290. We identified 12 publications that reported 26 adjusted arVE results. Five publications reported 13 laboratory confirmed arVE and seven reported 13 code-ascertained arVE. Nine publications with 22 results were at low risk of bias. Heterogeneity was explained by season. We found a significant 11% (8 to 14%) adjusted arVE favoring ccIV in preventing any IRO in the 2017–2018 influenza season. The arVE was 3% (−2% to 7%) in the 2018–2019 influenza season. We found moderate evidence of a significant advantage of the ccIV in preventing IRO, compared to eIV, in a well-matched A(H3N2) predominant season.
Collapse
|
32
|
Peck H, Laurie KL, Rockman S, Leung V, Lau H, Soppe S, Rynehart C, Baas C, Trusheim H, Barr IG. Enhanced isolation of influenza viruses in qualified cells improves the probability of well-matched vaccines. NPJ Vaccines 2021; 6:149. [PMID: 34887440 PMCID: PMC8660794 DOI: 10.1038/s41541-021-00415-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/12/2021] [Indexed: 01/04/2023] Open
Abstract
Influenza vaccines are utilised to combat seasonal and pandemic influenza. The key to influenza vaccination currently is the availability of candidate vaccine viruses (CVVs). Ideally, CVVs reflect the antigenic characteristics of the circulating virus, which may vary depending upon the isolation method. For traditional inactivated egg-based vaccines, CVVs are isolated in embryonated chicken eggs, while for cell-culture production, CVV's are isolated in either embryonated eggs or qualified cell lines. We compared isolation rates, growth characteristics, genetic stability and antigenicity of cell and egg CVV's derived from the same influenza-positive human clinical respiratory samples collected from 2008-2020. Influenza virus isolation rates in MDCK33016PF cells were twice that of eggs and mutations in the HA protein were common in egg CVVs but rare in cell CVVs. These results indicate that fully cell-based influenza vaccines will improve the choice, match and potentially the effectiveness, of seasonal influenza vaccines compared to egg-based vaccines.
Collapse
Affiliation(s)
- Heidi Peck
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia.
| | | | - Steve Rockman
- Seqirus Ltd, Parkville, VIC, Australia.,Department of Immunology and Microbiology, The University of Melbourne, Parkville, VIC, Australia
| | - Vivian Leung
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Hilda Lau
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Sally Soppe
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Cleve Rynehart
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | | | | | - Ian G Barr
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia.,Department of Immunology and Microbiology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
33
|
Chanthavanich P, Versage E, Van Twuijver E, Hohenboken M. Antibody responses against heterologous A/H5N1 strains for an MF59-adjuvanted cell culture-derived A/H5N1 (aH5N1c) influenza vaccine in healthy pediatric subjects. Vaccine 2021; 39:6930-6935. [PMID: 34711436 DOI: 10.1016/j.vaccine.2021.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/02/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Vaccines are the main prophylactic measure against pandemic influenza. Adjuvanted, cell culture-derived vaccines, which are not subject to limitations of egg-based vaccine production, have the potential to elicit an antibody response against heterologous strains and may be beneficial in the event of an A/H5N1 pandemic. METHODS A prespecified exploratory analysis of data from a phase 2, randomized, controlled, observer-blind multicenter trial (NCT01776554) to evaluate the immunogenicity of a MF59-adjuvanted, cell culture-based A/H5N1 influenza vaccine (aH5N1c), containing 7.5 µg hemagglutinin antigen per dose, in subjects 6 months through 17 years of age was conducted. Geometric mean titers (GMT) were determined using hemagglutination inhibition (HI) and microneutralization (MN) assays, and proportions of patients achieving seroconversion, HI and MN titers ≥ 1:40, and a 4-fold increase in MN titers against 5 heterologous strains (influenza A/H5N1 Anhui/2005, Egypt/2010, Hubei/2010, Indonesia/2005, and Vietnam/1203/2004) three weeks after administration of the second dose were assessed. RESULTS After the second dose, HI GMTs against heterologous strains increased between 8- and 40-fold, and MN GMTs increased 13- to 160-fold on Day 43 vs Day 1. On Day 43, 32-72% of subjects had HI titers ≥ 1:40 and achieved seroconversion against the heterologous strains. Using the MN assay, 84-100% of subjects had MN titers ≥ 1:40 and 83-100% achieved an at least 4-fold increase in MN titers against the heterologous strains. The highest responses were consistently against A/H5N1 Egypt/2010. CONCLUSIONS When given to children aged 6 months through 17 years, aH5N1c resulted in increased immunogenicity from baseline against all 5 heterologous A/H5N1 strains tested, demonstrating the potential of an MF59-adjuvanted, cell-derived A/H5N1 vaccine to provide cross-protection against other A/H5N1 strains (NCT01776554).
Collapse
Affiliation(s)
- Pornthep Chanthavanich
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Eve Versage
- Seqirus Inc., Clinical Development, Cambridge, USA
| | | | | |
Collapse
|
34
|
Transcriptomic Characterization Reveals Attributes of High Influenza Virus Productivity in MDCK Cells. Viruses 2021; 13:v13112200. [PMID: 34835006 PMCID: PMC8620111 DOI: 10.3390/v13112200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
The Madin–Darby Canine Kidney (MDCK) cell line is among the most commonly used cell lines for the production of influenza virus vaccines. As cell culture-based manufacturing is poised to replace egg-based processes, increasing virus production is of paramount importance. To shed light on factors affecting virus productivity, we isolated a subline, H1, which had twice the influenza virus A (IAV) productivity of the parent (P) through cell cloning, and characterized H1 and P in detail on both physical and molecular levels. Transcriptome analysis revealed that within a few hours after IAV infection, viral mRNAs constituted over one fifth of total mRNA, with several viral genes more highly expressed in H1 than P. Functional analysis of the transcriptome dynamics showed that H1 and P responded similarly to IAV infection, and were both subjected to host shutoff and inflammatory responses. Importantly, H1 was more active in translation and RNA processing intrinsically and after infection. Furthermore, H1 had more subdued inflammatory and antiviral responses. Taken together, we postulate that the high productivity of IAV hinges on the balance between suppression of host functions to divert cellular resources and the sustaining of sufficient activities for virus replication. Mechanistic insights into virus productivity can facilitate the process optimization and cell line engineering for advancing influenza vaccine manufacturing.
Collapse
|
35
|
Ortiz de Lejarazu-Leonardo R, Montomoli E, Wojcik R, Christopher S, Mosnier A, Pariani E, Trilla Garcia A, Fickenscher H, Gärtner BC, Jandhyala R, Zambon M, Moore C. Estimation of Reduction in Influenza Vaccine Effectiveness Due to Egg-Adaptation Changes-Systematic Literature Review and Expert Consensus. Vaccines (Basel) 2021; 9:1255. [PMID: 34835186 PMCID: PMC8621612 DOI: 10.3390/vaccines9111255] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Influenza vaccines are the main tool to prevent morbidity and mortality of the disease; however, egg adaptations associated with the choice of the manufacturing process may reduce their effectiveness. This study aimed to estimate the impact of egg adaptations and antigenic drift on the effectiveness of trivalent (TIV) and quadrivalent (QIV) influenza vaccines. METHODS Nine experts in influenza virology were recruited into a Delphi-style exercise. In the first round, the experts were asked to answer questions on the impact of antigenic drift and egg adaptations on vaccine match (VM) and influenza vaccine effectiveness (IVE). In the second round, the experts were presented with the data from a systematic literature review on the same subject and aggregated experts' responses to round one questions. The experts were asked to review and confirm or amend their responses before the final summary statistics were calculated. RESULTS The experts estimated that, across Europe, the egg adaptations reduce, on average, VM to circulating viruses by 7-21% and reduce IVE by 4-16%. According to the experts, antigenic drift results in a similar impact on VM (8-24%) and IVE (5-20%). The highest reduction in IVE was estimated for the influenza virus A(H3N2) subtype for the under 65 age group. When asked about the frequency of the phenomena, the experts indicated that, on average, between the 2014 and 19 seasons, egg adaptation and antigenic drift were significant enough to impact IVE that occurred in two and three out of five seasons, respectively. They also agreed that this pattern is likely to reoccur in future seasons. CONCLUSIONS Expert estimates suggest there is a potential for 9% on average (weighted average of "All strains" over three age groups adjusted by population size) and up to a 16% increase in IVE (against A(H3N2), the <65 age group) if egg adaptations that arise when employing the traditional egg-based manufacturing process are avoided.
Collapse
Affiliation(s)
| | - Emanuele Montomoli
- Department of Molecular Medicine, University of Siena, 53100 Siena, Italy;
| | - Radek Wojcik
- Medialis Ltd., Banbury OX16 0AH, UK; (S.C.); (R.J.)
| | | | | | - Elena Pariani
- Department of Biomedical Science for Health, University of Milan, 20122 Milan, Italy;
| | - Antoni Trilla Garcia
- Preventive Medicine and Epidemiology, Hospital Clínic, University of Barcelona, 08007 Barcelona, Spain;
| | - Helmut Fickenscher
- Institute for Infection Medicine, Kiel University, 24118 Kiel, Germany;
- University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Barbara C. Gärtner
- Institute for Microbiology and Hygiene, Saarland University, Faculty of Medicine and Medical Center, Building 43, 66421 Homburg/Saar, Germany;
| | | | | | - Catherine Moore
- Wales Specialist Virology Centre, Public Health Wales, Cardiff CF14 4XW, UK;
| |
Collapse
|
36
|
Nolan T, Fortanier AC, Leav B, Põder A, Bravo LC, Szymański HT, Heeringa M, Vermeulen W, Matassa V, Smolenov I, Edelman JM. Efficacy of a Cell-Culture-Derived Quadrivalent Influenza Vaccine in Children. N Engl J Med 2021; 385:1485-1495. [PMID: 34644472 DOI: 10.1056/nejmoa2024848] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cell-culture-derived influenza vaccines may enable a closer antigenic match to circulating strains of influenza virus by avoiding egg-adapted mutations. METHODS We evaluated the efficacy of a cell-culture-derived quadrivalent inactivated influenza vaccine (IIV4c) using a Madin-Darby canine kidney cell line in children and adolescents 2 to less than 18 years of age. During three influenza seasons, participants from eight countries were enrolled in an observer-blinded, randomized clinical trial comparing IIV4c with a noninfluenza vaccine (meningococcal ACWY). All the participants received a dose of a trial vaccine. Children 2 to less than 9 years of age without previous influenza vaccination who were assigned to the IIV4c group received a second dose on day 29; their counterparts who were assigned to the comparator group received placebo. Participants were followed for at least 180 days for efficacy and safety. The presence of influenza virus in nasopharyngeal swabs from participants with influenza-like illness was confirmed by reverse-transcriptase-polymerase-chain-reaction assay and viral culture. A Cox proportional-hazards model was used to evaluate the efficacy of IIV4c as measured by the first occurrence of laboratory-confirmed type A or B influenza (primary end point). RESULTS Between 2017 and 2019, a total of 4514 participants were randomly assigned to receive IIV4c or the meningococcal ACWY vaccine. Laboratory-confirmed influenza occurred in 175 of 2257 participants (7.8%) in the IIV4c group and in 364 of 2252 participants (16.2%) in the comparator group, and the efficacy of IIV4c was 54.6% (95% confidence interval [CI], 45.7 to 62.1). Efficacy was 80.7% (95% CI, 69.2 to 87.9) against influenza A/H1N1, 42.1% (95% CI, 20.3 to 57.9) against influenza A/H3N2, and 47.6% (95% CI, 31.4 to 60.0) against influenza B. IIV4c showed consistent vaccine efficacy in subgroups according to age, sex, race, and previous influenza vaccination. The incidences of adverse events were similar in the IIV4c group and the comparator group. CONCLUSIONS IIV4c provided protection against influenza in healthy children and adolescents across seasons, regardless of previous influenza vaccination. (Funded by Seqirus; EudraCT number, 2016-002883-15; ClinicalTrials.gov number, NCT03165617.).
Collapse
Affiliation(s)
- Terence Nolan
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Alexandre C Fortanier
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Brett Leav
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Airi Põder
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Lulu C Bravo
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Henryk T Szymański
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Marten Heeringa
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Wim Vermeulen
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Vince Matassa
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Igor Smolenov
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| | - Jonathan M Edelman
- From the Murdoch Children's Research Institute and the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne (T.N.), and Seqirus Clinical Development, Parkville (V.M.) - both in VIC, Australia; Seqirus Clinical Development, Amsterdam (A.C.F., M.H., W.V.); Seqirus Clinical Development, Cambridge, MA (B.L., I.S., J.M.E.); the Clinical Research Center, Tartu, Estonia (A.P.); the University of the Philippines, Manila (L.C.B.); and Hedwig of Silesia Hospital, Trzebnica, Poland (H.T.S.)
| |
Collapse
|
37
|
Bansal A, Trieu MC, Mohn KGI, Cox RJ. Safety, Immunogenicity, Efficacy and Effectiveness of Inactivated Influenza Vaccines in Healthy Pregnant Women and Children Under 5 Years: An Evidence-Based Clinical Review. Front Immunol 2021; 12:744774. [PMID: 34691051 PMCID: PMC8526930 DOI: 10.3389/fimmu.2021.744774] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/08/2021] [Indexed: 02/03/2023] Open
Abstract
Annual influenza vaccination is often recommended for pregnant women and young children to reduce the risk of severe influenza. However, most studies investigating the safety, immunogenicity, and efficacy or effectiveness of influenza vaccines are conducted in healthy adults. In this evidence-based clinical review, we provide an update on the safety profile, immunogenicity, and efficacy/effectiveness of inactivated influenza vaccines (IIVs) in healthy pregnant women and children <5 years old. Six electronic databases were searched until May 27, 2021. We identified 3,731 articles, of which 93 met the eligibility criteria and were included. The IIVs were generally well tolerated in pregnant women and young children, with low frequencies of adverse events following IIV administration; however, continuous vaccine safety monitoring systems are necessary to detect rare adverse events. IIVs generated good antibody responses, and the seroprotection rates after IIVs were moderate to high in pregnant women (range = 65%-96%) and young children (range = 50%-100%), varying between the different influenza types/subtypes and seasons. Studies show vaccine efficacy/effectiveness values of 50%-70% in pregnant women and 20%-90% in young children against lab-confirmed influenza, although the efficacy/effectiveness depended on the study design, host factors, vaccine type, manufacturing practices, and the antigenic match/mismatch between the influenza vaccine strains and the circulating strains. Current evidence suggests that the benefits of IIVs far outweigh the potential risks and that IIVs should be recommended for pregnant women and young children.
Collapse
Affiliation(s)
- Amit Bansal
- The Influenza Centre, Department of Clinical Sciences, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Mai-Chi Trieu
- The Influenza Centre, Department of Clinical Sciences, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kristin G I Mohn
- The Influenza Centre, Department of Clinical Sciences, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Rebecca Jane Cox
- The Influenza Centre, Department of Clinical Sciences, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Helse Bergen, Bergen, Norway
| |
Collapse
|
38
|
Bartley JM, Cadar AN, Martin DE. Better, Faster, Stronger: mRNA Vaccines Show Promise for Influenza Vaccination in Older Adults. Immunol Invest 2021; 50:810-820. [PMID: 33830864 DOI: 10.1080/08820139.2021.1909617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Older adults have diminished immune responses that lead to increased susceptibility and severity of infectious diseases. Influenza is a leading killer of older adults despite the availability of seasonal influenza vaccination. Influenza vaccines are strain specific, and their efficacy varies greatly year to year based on how well the vaccine virus matches the circulating strains. Additionally, older adults have reduced vaccination responses. The COVID-19 pandemic highlighted the increased mortality rate in older adults for infectious disease, and brought vaccine development to the forefront. The speed of vaccine development was met with an equally impressive vaccine efficacy. Interestingly, both mRNA-based COVID-19 vaccines currently available have shown similar efficacy in both young and older adults. mRNA vaccine production has significantly reduced the production timeline compared to current influenza vaccines, making them particularly attractive for influenza vaccine development. Faster production coupled with improved efficacy would be a tremendous advancement in protecting older adults from influenza morbidity and mortality.
Collapse
Affiliation(s)
- Jenna M Bartley
- Center on Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Andreia N Cadar
- Center on Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Dominique E Martin
- Center on Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
39
|
de Lusignan S, Tsang RSM, Amirthalingam G, Akinyemi O, Sherlock J, Tripathy M, Deeks A, Ferreira F, Howsam G, Hobbs FDR, Joy M. Adverse events of interest following influenza vaccination, a comparison of cell culture-based with egg-based alternatives: English sentinel network annual report paper 2019/20. LANCET REGIONAL HEALTH-EUROPE 2021; 2:100029. [PMID: 34557791 PMCID: PMC8454842 DOI: 10.1016/j.lanepe.2021.100029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background The cell-based quadrivalent influenza vaccine (QIVc) is now offered as an alternative to egg-based quadrivalent (QIVe) and adjuvanted trivalent (aTIV) influenza vaccines in the UK. While post-licensure studies show non-inferiority of cell-based vaccines, it is not known how its safety profile compares to other types of vaccines in real-world use. Methods We conducted a retrospective cohort study using computerised medical records from the Royal College of General Practitioners (RCGP) Research and Surveillance Centre (RSC) sentinel network database. We used a self-controlled case series design and calculated the relative incidence (RI) of adverse events of interest (AEIs) over different risk periods. We then compared the RIs of AEIs within seven days of vaccination overall and between QIVc and QIVe in the 18–64 years age group, and between QIVc and aTIV in the ≥65 years age group. Findings The majority of AEIs occurred within seven days of vaccination, and a seasonal effect was observed. Using QIVc as the reference group, QIVe showed similar incidence of AEIs whereas live attenuated influenza vaccine (LAIV) and aTIV had lower incidence of AEIs. In the stratified analyses, QIVe and aTIV were associated with a 16% lower incidence of AEIs in the seven days post-vaccination in both the 18–64 years and ≥65 years age groups. Interpretation Routine sentinel network data allow comparisons of safety profiles of equally suitable seasonal influenza vaccines. The higher incidence of AEIs associated with QIVc suggest monitoring of several seasons would allow robust comparisons to be made. Funding Public Health England.
Collapse
Affiliation(s)
- Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom.,Royal College of General Practitioners Research and Surveillance Centre, 30 Euston Square, London NW1 2FB, United Kingdom
| | - Ruby S M Tsang
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | | | - Oluwafunmi Akinyemi
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Julian Sherlock
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Manasa Tripathy
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Alexandra Deeks
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Filipa Ferreira
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Gary Howsam
- Royal College of General Practitioners Research and Surveillance Centre, 30 Euston Square, London NW1 2FB, United Kingdom
| | - F D Richard Hobbs
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| | - Mark Joy
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Woodstock Road, Oxford OX2 6GG, United Kingdom
| |
Collapse
|
40
|
Isakova-Sivak I, Stepanova E, Mezhenskaya D, Matyushenko V, Prokopenko P, Sychev I, Wong PF, Rudenko L. Influenza vaccine: progress in a vaccine that elicits a broad immune response. Expert Rev Vaccines 2021; 20:1097-1112. [PMID: 34348561 DOI: 10.1080/14760584.2021.1964961] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The licensed seasonal influenza vaccines predominantly induce neutralizing antibodies against immunodominant hypervariable epitopes of viral surface proteins, with limited protection against antigenically distant influenza viruses. Strategies have been developed to improve vaccines' performance in terms of broadly reactive and long-lasting immune response induction. AREAS COVERED We have summarized the advancements in the development of cross-protective influenza vaccines and discussed the challenges in evaluating them in preclinical and clinical trials. Here, the literature regarding the current stage of development of universal influenza vaccine candidates was reviewed. EXPERT OPINION Although various strategies aim to redirect adaptive immune responses from variable immunodominant to immunosubdominant antigens, more conserved epitopes are being investigated. Approaches that improve antibody responses to conserved B cell epitopes have increased the protective efficacy of vaccines within a subtype or phylogenetic group of influenza viruses. Vaccines that elicit significant levels of T cells recognizing highly conserved viral epitopes possess a high cross-protective potential and may cover most circulating influenza viruses. However, the development of T cell-based universal influenza vaccines is challenging owing to the diversity of MHCs in the population, unpredictable degree of immunodominance, lack of adequate animal models, and difficulty in establishing T cell immunity in humans. ABBREVIATIONS cHA: chimeric HA; HBc: hepatitis B virus core protein; HA: hemagglutinin; HLA: human leucocyte antigen; IIV: inactivated influenza vaccine; KLH: keyhole limpet hemocyanin; LAH: long alpha helix; LAIV: live attenuated influenza vaccine; M2e: extracellular domain of matrix 2 protein; MHC: major histocompatibility complex; mRNA: messenger ribonucleic acid; NA: neuraminidase; NS1: non-structural protein 1; qNIV: quadrivalent nanoparticle influenza vaccine; TRM: tissue-resident memory T cells; VE: vaccine effectiveness; VLP: virus-like particles; VSV: vesicular stomatitis virus.
Collapse
Affiliation(s)
- Irina Isakova-Sivak
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| | - Ekaterina Stepanova
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| | - Daria Mezhenskaya
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| | - Victoria Matyushenko
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| | - Polina Prokopenko
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| | - Ivan Sychev
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| | - Pei-Fong Wong
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| | - Larisa Rudenko
- Department Of Virology, Institute Of Experimental Medicine, Saint Petersburg, Russia
| |
Collapse
|
41
|
Boikos C, Fischer L, O'Brien D, Vasey J, Sylvester GC, Mansi JA. Relative Effectiveness of the Cell-derived Inactivated Quadrivalent Influenza Vaccine Versus Egg-derived Inactivated Quadrivalent Influenza Vaccines in Preventing Influenza-related Medical Encounters During the 2018-2019 Influenza Season in the United States. Clin Infect Dis 2021; 73:e692-e698. [PMID: 33400775 PMCID: PMC8326580 DOI: 10.1093/cid/ciaa1944] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/01/2021] [Indexed: 12/13/2022] Open
Abstract
Background The cell-propagated inactivated quadrivalent influenza vaccine (ccIIV4) may offer improved protection in seasons where egg-derived influenza viruses undergo mutations that affect antigenicity. This study estimated the relative vaccine effectiveness (rVE) of ccIIV4 versus egg-derived inactivated quadrivalent influenza vaccine (eIIV4) in preventing influenza-related medical encounters in the 2018–2019 US season. Methods A dataset linking primary care electronic medical records with medical claims data was used to conduct a retrospective cohort study among individuals ≥ 4 years old vaccinated with ccIIV4 or eIIV4 during the 2018–2019 season. Adjusted odds ratios (ORs) were derived from a doubly robust inverse probability of treatment-weighted approach adjusting for age, sex, race, ethnicity, geographic region, vaccination week, and health status. rVE was estimated by (1 – OR) × 100 and presented with 95% confidence intervals (CI). Results Following the application of inclusion/exclusion criteria, the study cohort included 2 125 430 ccIIV4 and 8 000 903 eIIV4 recipients. Adjusted analyses demonstrated a greater reduction in influenza-related medical encounters with ccIIV4 versus eIIV4, with the following rVE: overall, 7.6% (95% CI, 6.5–8.6); age 4–17 years, 3.9% (95% CI, .9–7.0); 18–64 years, 6.5% (95% CI, 5.2–7.9); 18–49 years, 7.5% (95% CI, 5.7–9.3); 50–64 years, 5.6% (95% CI, 3.6–7.6); and ≥65 years, –2.2% (95% CI, –5.4 to .9). Conclusions Adjusted analyses demonstrated statistically significantly greater reduction in influenza-related medical encounters in individuals vaccinated with ccIIV4 versus eIIV4 in the 2018–2019 US influenza season. These results support ccIIV4 as a potentially more effective public health measure against influenza than an egg-based equivalent.
Collapse
Affiliation(s)
| | | | | | - Joe Vasey
- Veradigm, San Francisco, California, USA
| | | | | |
Collapse
|
42
|
Kwetkat A, Heppner HJ, Endres AS, Leischker A. [Influenza]. Internist (Berl) 2021; 62:801-806. [PMID: 34236439 DOI: 10.1007/s00108-021-01101-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Influenza is the infectious disease with the highest population-based mortality. It mainly affects those aged 60 years and older, mainly due to immune senescence, which also favors complicated courses and compromises vaccine effectiveness. Therefore, various approaches have been developed for more immunogenic vaccines, which are now available for use. The Ständige Impfkommission (STIKO) has taken this into account in its current recommendation on influenza vaccination and has recommended a quadrivalent, inactivated high-dose vaccine as the standard vaccine for all ≥ 60-year-olds. Despite these successes, vaccination for prevention remains underutilized. Germany has never reached the WHO vaccination target of 75% of the elderly population. The main reasons for this are a lack of confidence in the effectiveness of vaccination, a lack of/restricted risk perception of the disease, and barriers to implementation. Initial approaches to overcoming these barriers, such as low-threshold vaccination services by involving pharmacies, are being implemented. However, further steps are needed to realize the potential of influenza vaccination, especially for such vulnerable groups as older adults.
Collapse
Affiliation(s)
- Anja Kwetkat
- Klinik für Geriatrie, Universitätsklinikum Jena, Bachstraße 18, 07743, Jena, Deutschland.
| | - Hans Jürgen Heppner
- Klinik für Geriatrie, Helios Klinikum Schwelm, Schwelm, Deutschland.,Lehrstuhl für Geriatrie, Universität Witten-Herdecke, Witten, Deutschland
| | | | - Andreas Leischker
- Klinik für Geriatrie, Alexianer Krefeld GmbH, Krefeld, Deutschland.,Klinik für Geriatrische Rehabilitation, Alexianer Tönisvorst GmbH, Tönisvorst, Deutschland
| |
Collapse
|
43
|
Bissinger T, Wu Y, Marichal-Gallardo P, Riedel D, Liu X, Genzel Y, Tan WS, Reichl U. Towards integrated production of an influenza A vaccine candidate with MDCK suspension cells. Biotechnol Bioeng 2021; 118:3996-4013. [PMID: 34219217 DOI: 10.1002/bit.27876] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/01/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022]
Abstract
Seasonal influenza epidemics occur both in northern and southern hemispheres every year. Despite the differences in influenza virus surface antigens and virulence of seasonal subtypes, manufacturers are well-adapted to respond to this periodical vaccine demand. Due to decades of influenza virus research, the development of new influenza vaccines is relatively straight forward. In similarity with the ongoing coronavirus disease 2019 pandemic, vaccine manufacturing is a major bottleneck for a rapid supply of the billions of doses required worldwide. In particular, egg-based vaccine production would be difficult to schedule and shortages of other egg-based vaccines with high demands also have to be anticipated. Cell culture-based production systems enable the manufacturing of large amounts of vaccines within a short time frame and expand significantly our options to respond to pandemics and emerging viral diseases. In this study, we present an integrated process for the production of inactivated influenza A virus vaccines based on a Madin-Darby Canine Kidney (MDCK) suspension cell line cultivated in a chemically defined medium. Very high titers of 3.6 log10 (HAU/100 µl) were achieved using fast-growing MDCK cells at concentrations up to 9.5 × 106 cells/ml infected with influenza A/PR/8/34 H1N1 virus in 1 L stirred tank bioreactors. A combination of membrane-based steric-exclusion chromatography followed by pseudo-affinity chromatography with a sulfated cellulose membrane adsorber enabled full recovery for the virus capture step and up to 80% recovery for the virus polishing step. Purified virus particles showed a homogenous size distribution with a mean diameter of 80 nm. Based on a monovalent dose of 15 µg hemagglutinin (single-radial immunodiffusion assay), the level of total protein and host cell DNA was 58 µg and 10 ng, respectively. Furthermore, all process steps can be fully scaled up to industrial quantities for commercial manufacturing of either seasonal or pandemic influenza virus vaccines. Fast production of up to 300 vaccine doses per liter within 4-5 days makes this process competitive not only to other cell-based processes but to egg-based processes as well.
Collapse
Affiliation(s)
- Thomas Bissinger
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Yixiao Wu
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Pavel Marichal-Gallardo
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Dietmar Riedel
- Facility for Transmission Electron Microscopy, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Xuping Liu
- Shanghai BioEngine Sci-Tech Co., Shanghai, China
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.,Shanghai BioEngine Sci-Tech Co., Shanghai, China
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,Chair of Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
44
|
Ramjee L, Lemay W, Vurgun N, Charland N, Bauch CT, Pullagura GR, Houle SKD, Tremblay G. Projected impact of a plant-derived vaccine on the burden of seasonal influenza in Canada. Hum Vaccin Immunother 2021; 17:3643-3651. [PMID: 34213404 PMCID: PMC8437550 DOI: 10.1080/21645515.2021.1908797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective The analysis estimates projected population outcomes resulting from the introduction of a plant-derived influenza vaccine formulated as quadrivalent virus-like particles (QVLP) in Canada. Methods Using Monte Carlo simulations, the number of influenza cases, general practitioner visits, inpatient admissions, intensive care unit (ICU) admissions, and deaths due to influenza-associated illness were estimated under no vaccination, plant-derived QVLP vaccines only, or egg-derived vaccines only. The base case analysis examined the adult Canadian population in two subgroups: 18–64 years of age during the 2017/18 season and 65+ years of age during the 2018/19 season. Efficacy data were obtained from QVLP clinical trials. Vaccine effectiveness data for egg-derived vaccines were calculated from observational studies from the corresponding influenza seasons. Scenario analyses examined the impact of varying absolute vaccine effectiveness or vaccination coverage from base case inputs. Results In the base case analysis, plant-derived QVLP vaccines led to an additional reduction in the burden of influenza over egg-derived vaccines for both population subgroups. In the 18–64 subgroup, QVLP vaccines were associated with 2.63% (48,029; 95% credible interval [Crl]: 42,723–53,336) fewer influenza cases than egg-derived vaccines. In the 65+ subgroup, QVLP vaccines led to 4.82% (27,918; 95% Crl: 25,440–30,397) fewer influenza cases, and reductions in the number of inpatient admissions by 4.77% (1167; 95% CrI: 851–1483) and deaths by 4.75% (326; 95% CrI: 107–546) compared to egg-derived vaccines. Further reductions were observed in scenario analyses considering the potential increase in vaccine coverage. Conclusion Use of plant-derived QVLP influenza vaccines may contribute to greater reductions in influenza cases and influenza-related outcomes, including inpatient admissions and deaths, compared to egg-derived vaccines currently available in Canada.
Collapse
Affiliation(s)
- Lauren Ramjee
- Health Economics, Purple Squirrel Economics, Montréal, QC, Canada
| | - William Lemay
- Health Economics, Purple Squirrel Economics, Montréal, QC, Canada
| | - Nesrin Vurgun
- Health Economics, Purple Squirrel Economics, Montréal, QC, Canada
| | | | - Chris T Bauch
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON, Canada
| | | | | | - Gabriel Tremblay
- Health Economics, Purple Squirrel Economics, Montréal, QC, Canada
| |
Collapse
|
45
|
Martin ET, Cheng C, Petrie JG, Alyanak E, Gaglani M, Middleton DB, Ghamande S, Silveira FP, Murthy K, Zimmerman RK, Monto AS, Trabue C, Talbot HK, Ferdinands JM. Low Influenza Vaccine Effectiveness Against A(H3N2)-Associated Hospitalizations in 2016-2017 and 2017-2018 of the Hospitalized Adult Influenza Vaccine Effectiveness Network (HAIVEN). J Infect Dis 2021; 223:2062-2071. [PMID: 33140094 DOI: 10.1093/infdis/jiaa685] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/30/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The 2016-2017 and 2017-2018 influenza seasons were notable for the high number of hospitalizations for influenza A(H3N2) despite vaccine and circulating strain match. METHODS We evaluated vaccine effectiveness (VE) against hospitalization in the test-negative HAIVEN study. Nasal-throat swabs were tested by quantitative reverse transcription polymerase chain reaction (RT-PCR) for influenza and VE was determined based on odds of vaccination by generalized estimating equations. Vaccine-specific antibody was measured in a subset of enrollees. RESULTS A total of 6129 adults were enrolled from 10 hospitals. Adjusted VE against A(H3N2) was 22.8% (95% confidence interval [CI], 8.3% to 35.0%), pooled across both years and 49.4% (95% CI, 34.3% to 61.1%) against B/Yamagata. In 2017-2018, the A(H3N2) VE point estimate for the cell-based vaccine was 43.0% (95% CI, -36.3% to 76.1%; 56 vaccine recipients) compared to 24.0% (95% CI, 3.9% to 39.9%) for egg-based vaccines. Among 643 with serology data, hemagglutinin antibodies against the egg-based A(H3N2) vaccine strain were increased in influenza-negative individuals. CONCLUSIONS Low VE for the A/Hong Kong/4801/2014 vaccine virus in both A(H3N2) seasons emphasizes concerns for continued changes in H3N2 antigenic epitopes, including changes that may impact glycosylation and ultimately reduce VE.
Collapse
Affiliation(s)
- Emily T Martin
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Caroline Cheng
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Joshua G Petrie
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Elif Alyanak
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Manjusha Gaglani
- Baylor Scott and White Health, Texas A&M University Health Science Center College of Medicine, Temple, Texas, USA
| | | | - Shekhar Ghamande
- Baylor Scott and White Health, Texas A&M University Health Science Center College of Medicine, Temple, Texas, USA
| | | | | | | | - Arnold S Monto
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Christopher Trabue
- Ascension Saint Thomas, Nashville, Tennessee, USA.,Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - H Keipp Talbot
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jill M Ferdinands
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | |
Collapse
|
46
|
Lutz CS, Biggerstaff M, Rolfes MA, Lafond KE, Azziz-Baumgartner E, Porter RM, Reed C, Bresee JS. Estimating the number of averted illnesses and deaths as a result of vaccination against an influenza pandemic in nine low- and middle-income countries. Vaccine 2021; 39:4219-4230. [PMID: 34119348 DOI: 10.1016/j.vaccine.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND During the 2009 influenza A(H1N1)pdm09 pandemic, 77 countries received donated monovalent A(H1N1)pdm09 vaccine through the WHO Pandemic Influenza A(H1N1) Vaccine Deployment Initiative. However, 47% did not receive their first shipment until after the first wave of virus circulation, and 8% did not receive their first shipment until after the WHO declared the end of the pandemic. Arguably, these shipments were too late into the pandemic to have a substantial effect on virus transmission or disease burden during the first waves of the pandemic. OBJECTIVES In order to evaluate the potential benefits of earlier vaccine availability, we estimated the number of illnesses and deaths that could be averted during a 2009-like influenza pandemic under five different vaccine-availability timing scenarios. METHODS We adapted a model originally developed to estimate annual influenza morbidity and mortality burden averted through US seasonal vaccination and ran it for five vaccine availability timing scenarios in nine low- and middle-income countries that received donated vaccine. RESULTS Among nine study countries, we estimated that the number of averted cases was 61-216,197 for actual vaccine receipt, increasing to 2,914-283,916 had vaccine been available simultaneously with the United States. CONCLUSIONS Earlier delivery of vaccines can reduce influenza case counts during a simulated 2009-like pandemic in some low- and middle-income countries. For others, increasing the number of cases and deaths prevented through vaccination may be dependent on factors other than timely initiation of vaccine administration, such as distribution and administration capacity.
Collapse
Affiliation(s)
- Chelsea S Lutz
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States; Oak Ridge Institute for Science and Education, United States Department of Energy, 100 ORAU Way, Oak Ridge, TN 37830, United States; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States.
| | - Matthew Biggerstaff
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Melissa A Rolfes
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Kathryn E Lafond
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Eduardo Azziz-Baumgartner
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Rachael M Porter
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Carrie Reed
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| | - Joseph S Bresee
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Rd NE, Atlanta, GA 30329, United States
| |
Collapse
|
47
|
Sekiya T, Ohno M, Nomura N, Handabile C, Shingai M, Jackson DC, Brown LE, Kida H. Selecting and Using the Appropriate Influenza Vaccine for Each Individual. Viruses 2021; 13:971. [PMID: 34073843 PMCID: PMC8225103 DOI: 10.3390/v13060971] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022] Open
Abstract
Despite seasonal influenza vaccines having been routinely used for many decades, influenza A virus continues to pose a global threat to humans, causing high morbidity and mortality each year. The effectiveness of the vaccine is largely dependent on how well matched the vaccine strains are with the circulating influenza virus strains. Furthermore, low vaccine efficacy in naïve populations such as young children, or in the elderly, who possess weakened immune systems, indicates that influenza vaccines need to be more personalized to provide broader community protection. Advances in both vaccine technologies and our understanding of influenza virus infection and immunity have led to the design of a variety of alternate vaccine strategies to extend population protection against influenza, some of which are now in use. In this review, we summarize the progress in the field of influenza vaccines, including the advantages and disadvantages of different strategies, and discuss future prospects. We also highlight some of the challenges to be faced in the ongoing effort to control influenza through vaccination.
Collapse
Affiliation(s)
- Toshiki Sekiya
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia
| | - Marumi Ohno
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
| | - Naoki Nomura
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
| | - Chimuka Handabile
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
| | - Masashi Shingai
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
| | - David C. Jackson
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia
| | - Lorena E. Brown
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia
| | - Hiroshi Kida
- International Institute for Zoonosis Control, Hokkaido University, Kita-20 Nishi-10, Kita-ku, Sapporo 001-0020, Japan; (T.S.); (M.O.); (N.N.); (C.H.); (M.S.)
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan; (D.C.J.); (L.E.B.)
- Collaborating Research Center for the Control of Infectious Diseases, Nagasaki University, Nagasaki 852-8521, Japan
| |
Collapse
|
48
|
A new and simplified anion exchange chromatographic process for the purification of cell-grown influenza A H1N1 virus. Sep Purif Technol 2021. [DOI: 10.1016/j.seppur.2021.118412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
49
|
Newland M, Durham D, Asher J, Treanor JJ, Seals J, Donis RO, Johnson RA. Improving pandemic preparedness through better, faster influenza vaccines. Expert Rev Vaccines 2021; 20:235-242. [PMID: 33576708 DOI: 10.1080/14760584.2021.1886931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction. Timely availability of effective influenza vaccine will be critical to mitigate the next influenza pandemic. The mission of Biomedical Advanced Research and Development Authority (BARDA) is to develop medical countermeasures against pandemics, including influenza and other health security threats.Areas covered. Despite considerable gains in pandemic vaccine preparedness since 2009, old and new challenges threaten the pandemic influenza response capabilities of the U.S. Government: insufficient U.S.-based vaccine production, two-dose vaccination regimen, logistically complex adjuvanted formulation, and sustained surge manufacturing capacity despite no commercial market for pandemic vaccines. Although the coronavirus disease 2019 (COVID-19) pandemic has re-exposed these gaps in preparedness and response, previous investments into flexible influenza vaccine technologies proved to be critical to accelerate COVID-19 vaccine development.Expert opinion. BARDA addresses these challenges by implementing a pandemic influenza vaccine strategy with two key goals: 1) accelerating vaccine development and production (faster) and 2) improving vaccine performance (better). This strategy involves an end-to-end approach, including increasing manufacturing and fill-finish capacity; improving release testing speed; and funding clinical trials to improve current vaccine utilization. As demonstrated by the COVID-19 response, continued investments into this pandemic influenza vaccine strategy will further enhance the ability to respond to future emerging pandemic pathogens.
Collapse
Affiliation(s)
- Matthew Newland
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington DC, USA
| | - David Durham
- Leidos Supporting the Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington DC, USA
| | - Jason Asher
- Leidos Supporting the Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington DC, USA
| | - John J Treanor
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington DC, USA
| | - Jonathan Seals
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington DC, USA
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington DC, USA
| | - Robert A Johnson
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington DC, USA
| |
Collapse
|
50
|
T Cell Immunity against Influenza: The Long Way from Animal Models Towards a Real-Life Universal Flu Vaccine. Viruses 2021; 13:v13020199. [PMID: 33525620 PMCID: PMC7911237 DOI: 10.3390/v13020199] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Current flu vaccines rely on the induction of strain-specific neutralizing antibodies, which leaves the population vulnerable to drifted seasonal or newly emerged pandemic strains. Therefore, universal flu vaccine approaches that induce broad immunity against conserved parts of influenza have top priority in research. Cross-reactive T cell responses, especially tissue-resident memory T cells in the respiratory tract, provide efficient heterologous immunity, and must therefore be a key component of universal flu vaccines. Here, we review recent findings about T cell-based flu immunity, with an emphasis on tissue-resident memory T cells in the respiratory tract of humans and different animal models. Furthermore, we provide an update on preclinical and clinical studies evaluating T cell-evoking flu vaccines, and discuss the implementation of T cell immunity in real-life vaccine policies.
Collapse
|