1
|
Zinnecker T, Reichl U, Genzel Y. Innovations in cell culture-based influenza vaccine manufacturing - from static cultures to high cell density cultivations. Hum Vaccin Immunother 2024; 20:2373521. [PMID: 39007904 PMCID: PMC11253887 DOI: 10.1080/21645515.2024.2373521] [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: 03/28/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Influenza remains a serious global health concern, causing significant morbidity and mortality each year. Vaccination is crucial to mitigate its impact, but requires rapid and efficient manufacturing strategies to handle timing and supply. Traditionally relying on egg-based production, the field has witnessed a paradigm shift toward cell culture-based methods offering enhanced flexibility, scalability, and process safety. This review provides a concise overview of available cell substrates and technological advancements. We summarize crucial steps toward process intensification - from roller bottle production to dynamic cultures on carriers and from suspension cultures in batch mode to high cell density perfusion using various cell retention devices. Moreover, we compare single-use and conventional systems and address challenges including defective interfering particles. Taken together, we describe the current state-of-the-art in cell culture-based influenza virus production to sustainably meet vaccine demands, guarantee a timely supply, and keep up with the challenges of seasonal epidemics and global pandemics.
Collapse
Affiliation(s)
- Tilia Zinnecker
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Bioprocess Engineering, Otto-von-Guericke University, Magdeburg, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| |
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
|
Luo J, Zhang M, Ye Q, Gao F, Xu W, Li B, Wang Q, Zhao L, Tan WS. A synthetic TLR4 agonist significantly increases humoral immune responses and the protective ability of an MDCK-cell-derived inactivated H7N9 vaccine in mice. Arch Virol 2024; 169:163. [PMID: 38990396 DOI: 10.1007/s00705-024-06082-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024]
Abstract
Antigenically divergent H7N9 viruses pose a potential threat to public health, with the poor immunogenicity of candidate H7N9 vaccines demonstrated in clinical trials underscoring the urgent need for more-effective H7N9 vaccines. In the present study, mice were immunized with various doses of a suspended-MDCK-cell-derived inactivated H7N9 vaccine, which was based on a low-pathogenic H7N9 virus, to assess cross-reactive immunity and cross-protection against antigenically divergent H7N9 viruses. We found that the CRX-527 adjuvant, a synthetic TLR4 agonist, significantly enhanced the humoral immune responses of the suspended-MDCK-cell-derived H7N9 vaccine, with significant antigen-sparing and immune-enhancing effects, including robust virus-specific IgG, hemagglutination-inhibiting (HI), neuraminidase-inhibiting (NI), and virus-neutralizing (VN) antibody responses, which are crucial for protection against influenza virus infection. Moreover, the CRX-527-adjuvanted H7N9 vaccine also elicited cross-protective immunity and cross-protection against a highly pathogenic H7N9 virus with a single vaccination. Notably, NI and VN antibodies might play an important role in cross-protection against lethal influenza virus infections. This study showed that a synthetic TLR4 agonist adjuvant has a potent immunopotentiating effect, which might be considered worth further development as a means of increasing vaccine effectiveness.
Collapse
Affiliation(s)
- Jian Luo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Institute of Biological Products, Shanghai, China
| | - Min Zhang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Qian Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Feixia Gao
- Shanghai Institute of Biological Products, Shanghai, China
| | - Wenting Xu
- Shanghai Institute of Biological Products, Shanghai, China
| | - Beibei Li
- Shanghai Institute of Biological Products, Shanghai, China
| | - Qi Wang
- Shanghai Institute of Biological Products, Shanghai, China
| | - Liang Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
4
|
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
|
5
|
Li F, Liu B, Xiong Y, Zhang Z, Zhang Q, Qiu R, Peng F, Nian X, Wu D, Li X, Liu J, Li Z, Tu H, Wu W, Wang Y, Zhang J, Yang X. Enhanced Downstream Processing for a Cell-Based Avian Influenza (H5N1) Vaccine. Vaccines (Basel) 2024; 12:138. [PMID: 38400122 PMCID: PMC10891636 DOI: 10.3390/vaccines12020138] [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: 11/24/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
H5N1 highly pathogenic avian influenza virus (HPAIV) infections pose a significant threat to human health, with a mortality rate of around 50%. Limited global approval of H5N1 HPAIV vaccines, excluding China, prompted the need to address safety concerns related to MDCK cell tumorigenicity. Our objective was to improve vaccine safety by minimizing residual DNA and host cell protein (HCP). We developed a downstream processing method for the cell-based H5N1 HPAIV vaccine, employing CaptoTM Core 700, a multimodal resin, for polishing. Hydrophobic-interaction chromatography (HIC) with polypropylene glycol as a functional group facilitated the reversible binding of virus particles for capture. Following the two-step chromatographic process, virus recovery reached 68.16%. Additionally, HCP and DNA levels were reduced to 2112.60 ng/mL and 6.4 ng/mL, respectively. Western blot, high-performance liquid chromatography (HPLC), and transmission electron microscopy (TEM) confirmed the presence of the required antigen with a spherical shape and appropriate particle size. Overall, our presented two-step downstream process demonstrates potential as an efficient and cost-effective platform technology for cell-based influenza (H5N1 HPAIV) vaccines.
Collapse
Affiliation(s)
- Fang Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Bo Liu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Yu Xiong
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zhegang Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Qingmei Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Ran Qiu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Feixia Peng
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Dongping Wu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuedan Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Jing Liu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Ze Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Hao Tu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Wenyi Wu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Yu Wang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China; (F.L.); (B.L.); (Y.X.); (Z.Z.); (Q.Z.); (R.Q.); (F.P.); (X.N.); (D.W.); (X.L.); (J.L.); (Z.L.); (H.T.); (W.W.); (Y.W.)
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research, Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
- China National Biotec Group Company Limited, Beijing 100029, China
| |
Collapse
|
6
|
Yang D, Huang L, Wang J, Wu H, Liu Z, Abudureyimu A, Qiao Z. Tumorigenesis mechanism and application strategy of the MDCK cell line: A systematic review. Biologicals 2023; 83:101699. [PMID: 37573790 DOI: 10.1016/j.biologicals.2023.101699] [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: 01/11/2023] [Revised: 06/26/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023] Open
Abstract
Influenza is an acute respiratory infectious disease caused by influenza virus that seriously endangers people's health. Influenza vaccination is the most effective means to prevent influenza virus infection and its serious complications. MDCK cells are considered to be superior to chicken embryos for the production of influenza vaccines, but the tumorigenicity of cells is a concern over the theoretical possibility of the risk of adverse events. The theoretical risks need to be adequately addressed if public confidence in programs of immunization are to be maintained. In this paper, studies of the tumorigenic potential of cell lines, with MDCK cells as an example, published since 2010 are reviewed. The mechanism of tumorigenicity of MDCK cells is discussed with reference to cell heterogeneity and epithelial to mesenchymal transition (EMT). Understanding the mechanism of the acquisition of a tumorigenic phenotype by MDCK cells might assist in estimating potential risks associated with using tumorigenic cell substrates for vaccine production.
Collapse
Affiliation(s)
- Di Yang
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, 730030, Lanzhou, China; College of Veterinary Medicine, Gansu Agricultural University, 730030, Lanzhou, China; Department of Experiment & Teaching, Northwest Minzu University, 730030, Lanzhou, China.
| | - Lingwei Huang
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, 730030, Lanzhou, China.
| | - Jiamin Wang
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, 730030, Lanzhou, China.
| | - Huihao Wu
- Department of Experiment & Teaching, Northwest Minzu University, 730030, Lanzhou, China.
| | - Zhenbin Liu
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, 730030, Lanzhou, China.
| | - Ayimuguli Abudureyimu
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, 730030, Lanzhou, China.
| | - Zilin Qiao
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, 730030, Lanzhou, China.
| |
Collapse
|
7
|
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
|
8
|
Trombetta CM, Marchi S, Montomoli E. The baculovirus expression vector system: a modern technology for the future of influenza vaccine manufacturing. Expert Rev Vaccines 2022; 21:1233-1242. [DOI: 10.1080/14760584.2022.2085565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
- VisMederi Research srl, Siena, Italy
| |
Collapse
|
9
|
Wang J, Liu L, Yang D, Zhang L, Abudureyimu A, Qiao Z, Ma Z. Identification and differential expression of microRNAs in Madin–Darby canine kidney cells with high and low tumorigenicities. Genes Genomics 2022; 44:187-196. [DOI: 10.1007/s13258-021-01177-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 10/12/2021] [Indexed: 11/24/2022]
|
10
|
Kim Y, Hong K, Kim H, Nam J. Influenza vaccines: Past, present, and future. Rev Med Virol 2022; 32:e2243. [PMID: 33949021 PMCID: PMC8209895 DOI: 10.1002/rmv.2243] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023]
Abstract
Globally, infection by seasonal influenza viruses causes 3-5 million cases of severe illness and 290,000-650,000 respiratory deaths each year. Various influenza vaccines, including inactivated split- and subunit-type, recombinant and live attenuated vaccines, have been developed since the 1930s when it was discovered that influenza viruses could be cultivated in embryonated eggs. However, the protection rate offered by these vaccines is rather low, especially in very young children and the elderly. In this review, we describe the history of influenza vaccine development, the immune responses induced by the vaccines and the adjuvants applied. Further, we suggest future directions for improving the effectiveness of influenza vaccines in all age groups. This includes the development of an influenza vaccine that induces a balanced T helper cell type 1 and type 2 immune responses based on the understanding of the immune system, and the development of a broad-spectrum influenza vaccine that can increase effectiveness despite antigen shifts and drifts, which are characteristics of the influenza virus. A brighter future can be envisaged if the development of an adjuvant that is safe and effective is realized.
Collapse
Affiliation(s)
- Yun‐Hee Kim
- Department of Medical and Biological SciencesThe Catholic University of KoreaBucheonRepublic of Korea
- Department of R&DSK BioscienceBundang‐guRepublic of Korea
| | - Kee‐Jong Hong
- UIC FoundationKonkuk UniversitySeoulRepublic of Korea
| | - Hun Kim
- Department of R&DSK BioscienceBundang‐guRepublic of Korea
| | - Jae‐Hwan Nam
- Department of Medical and Biological SciencesThe Catholic University of KoreaBucheonRepublic of Korea
| |
Collapse
|
11
|
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: 2.5] [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
|
12
|
Citiulo F, Crosatti C, Cattivelli L, Biselli C. Frontiers in the Standardization of the Plant Platform for High Scale Production of Vaccines. PLANTS (BASEL, SWITZERLAND) 2021; 10:1828. [PMID: 34579360 PMCID: PMC8467261 DOI: 10.3390/plants10091828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
The recent COVID-19 pandemic has highlighted the value of technologies that allow a fast setup and production of biopharmaceuticals in emergency situations. The plant factory system can provide a fast response to epidemics/pandemics. Thanks to their scalability and genome plasticity, plants represent advantageous platforms to produce vaccines. Plant systems imply less complicated production processes and quality controls with respect to mammalian and bacterial cells. The expression of vaccines in plants is based on transient or stable transformation systems and the recent progresses in genome editing techniques, based on the CRISPR/Cas method, allow the manipulation of DNA in an efficient, fast, and easy way by introducing specific modifications in specific sites of a genome. Nonetheless, CRISPR/Cas is far away from being fully exploited for vaccine expression in plants. In this review, an overview of the potential conjugation of the renewed vaccine technologies (i.e., virus-like particles-VLPs, and industrialization of the production process) with genome editing to produce vaccines in plants is reported, illustrating the potential advantages in the standardization of the plant platforms, with the overtaking of constancy of large-scale production challenges, facilitating regulatory requirements and expediting the release and commercialization of the vaccine products of genome edited plants.
Collapse
Affiliation(s)
- Francesco Citiulo
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy;
| | - Cristina Crosatti
- Council for Agricultural Research and Economics, Research Centre for Genomics and Bioinformatics, Via San Protaso 302, 29017 Fiorenzuola d’Arda, Italy; (C.C.); (L.C.)
| | - Luigi Cattivelli
- Council for Agricultural Research and Economics, Research Centre for Genomics and Bioinformatics, Via San Protaso 302, 29017 Fiorenzuola d’Arda, Italy; (C.C.); (L.C.)
| | - Chiara Biselli
- Council for Agricultural Research and Economics, Research Centre for Viticulture and Enology, Viale Santa Margherita 80, 52100 Arezzo, Italy
| |
Collapse
|
13
|
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: 13] [Impact Index Per Article: 3.3] [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
|
14
|
Boikos C, Sylvester GC, Sampalis JS, Mansi JA. Relative Effectiveness of the Cell-Cultured Quadrivalent Influenza Vaccine Compared to Standard, Egg-derived Quadrivalent Influenza Vaccines in Preventing Influenza-like Illness in 2017-2018. Clin Infect Dis 2021; 71:e665-e671. [PMID: 32253431 PMCID: PMC7745007 DOI: 10.1093/cid/ciaa371] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 04/03/2020] [Indexed: 11/29/2022] Open
Abstract
Background Influenza antigens may undergo adaptive mutations during egg-based vaccine production. In the 2017–2018 influenza season, quadrivalent, inactivated cell-derived influenza vaccine (ccIIV4) vaccine was produced using A(H3N2) seed virus propagated exclusively in cell culture, thus lacking egg adaptive changes. This United States study estimated relative vaccine effectiveness (rVE) of ccIIV4 vs egg-derived quadrivalent vaccines (egg-derived IIV4) for that season. Methods Vaccination, outcome, and covariate data were ascertained retrospectively from a electronic medical record (EMR) dataset and analyzed. The study cohort included patients ≥ 4 years of age. rVE was estimated against influenza-like illness (ILI) using diagnostic International Classification of Diseases, Ninth or Tenth Revision codes. The adjusted odds ratios used to derive rVE estimates were estimated from multivariable logistic regression models adjusted for age, sex, race/ethnicity, geographic region, and health status. Results Overall, 92 187 individuals had a primary care EMR record of ccIIV4 and 1 261 675 had a record of egg-derived IIV4. In the ccIIV4 group, 1705 narrowly defined ILI events occurred, and 25 645 occurred in the standard egg-derived IIV4 group. Crude rVE was 9.2% (95% confidence interval [CI], 4.6%–13.6%). When adjusted for age, sex, health status, comorbidities, and geographic region, the estimated rVE changed to 36.2% (95% CI, 26.1%–44.9%). Conclusions ccIIV4, derived from A(H3N2) seed virus propagated exclusively in cell culture, was more effective than egg-derived IIV4 in preventing ILI during the 2017–2018 influenza season. This result suggests that cell-derived influenza vaccines may have greater effectiveness than standard egg-derived vaccines.
Collapse
Affiliation(s)
| | | | - John S Sampalis
- Department of Experimental Surgery, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
15
|
Pech S, Rehberg M, Janke R, Benndorf D, Genzel Y, Muth T, Sickmann A, Rapp E, Reichl U. Tracking changes in adaptation to suspension growth for MDCK cells: cell growth correlates with levels of metabolites, enzymes and proteins. Appl Microbiol Biotechnol 2021; 105:1861-1874. [PMID: 33582836 PMCID: PMC7907048 DOI: 10.1007/s00253-021-11150-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 11/17/2022]
Abstract
Abstract Adaptations of animal cells to growth in suspension culture concern in particular viral vaccine production, where very specific aspects of virus-host cell interaction need to be taken into account to achieve high cell specific yields and overall process productivity. So far, the complexity of alterations on the metabolism, enzyme, and proteome level required for adaptation is only poorly understood. In this study, for the first time, we combined several complex analytical approaches with the aim to track cellular changes on different levels and to unravel interconnections and correlations. Therefore, a Madin-Darby canine kidney (MDCK) suspension cell line, adapted earlier to growth in suspension, was cultivated in a 1-L bioreactor. Cell concentrations and cell volumes, extracellular metabolite concentrations, and intracellular enzyme activities were determined. The experimental data set was used as the input for a segregated growth model that was already applied to describe the growth dynamics of the parental adherent cell line. In addition, the cellular proteome was analyzed by liquid chromatography coupled to tandem mass spectrometry using a label-free protein quantification method to unravel altered cellular processes for the suspension and the adherent cell line. Four regulatory mechanisms were identified as a response of the adaptation of adherent MDCK cells to growth in suspension. These regulatory mechanisms were linked to the proteins caveolin, cadherin-1, and pirin. Combining cell, metabolite, enzyme, and protein measurements with mathematical modeling generated a more holistic view on cellular processes involved in the adaptation of an adherent cell line to suspension growth. Key points • Less and more efficient glucose utilization for suspension cell growth • Concerted alteration of metabolic enzyme activity and protein expression • Protein candidates to interfere glycolytic activity in MDCK cells Supplementary Information The online version contains supplementary material available at 10.1007/s00253-021-11150-z.
Collapse
Affiliation(s)
- Sabine Pech
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Markus Rehberg
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Robert Janke
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Dirk Benndorf
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.
| | - Thilo Muth
- Section S.3 eScience, Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany.,Medizinische Fakultät, Medizinisches Proteom-Center (MPC), Ruhr-Universität Bochum, Bochum, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Erdmann Rapp
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,glyxera GmbH, Magdeburg, Germany
| | - Udo Reichl
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| |
Collapse
|
16
|
Ganguly M, Yeolekar L, Tyagi P, Sagar U, Narale S, Anaspure Y, Tupe S, Wadkar K, Ingle N, Dhere R, Scorza FB, Mahmood K. Evaluation of manufacturing feasibility and safety of an MDCK cell-based live attenuated influenza vaccine (LAIV) platform. Vaccine 2020; 38:8379-8386. [PMID: 33229107 DOI: 10.1016/j.vaccine.2020.10.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/24/2020] [Accepted: 10/31/2020] [Indexed: 12/23/2022]
Abstract
Cell culture based live attenuated influenza vaccines (LAIV) as an alternative to egg-based LAIV have been explored because of lack of easy access to SPF eggs for large scale production. In this study, feasibility of MDCK platform was assessed by including multiple LAIV strains covering both type A (H1 and H3) and type B seasonal strains as well as the candidate pandemic potential strains like A/H5 and A/H7 for the growth in MDCK cells. A risk assessment study was conducted on the cell banks to evaluate safety concerns related to tumorigenicity with a regulatory perspective. Tumorigenic potential of the MDCK cells was evaluated in nude mice (107cells/mouse) model system. The 50% tumor producing dose (TPD50) of MDCK cells was studied in SCID mice to determine the amount of cells required for induction of tumors. Further, we conducted an oncogenicity study in three sensitive rodent species as per the requirements specified in the WHO guidelines. We determined TPD50 value of 1.9 X 104 cells/mice through subcutaneous route. Our results suggest that, the intranasal route of administration of the cell culture based LAIV pose minimal to no risk of tumorigenicity associated with the host cells. Also, non-oncogenic nature of MDCK cells was demonstrated. Host cell DNA in the vaccine formulations was < 10 ng/dose which ensures vaccine safety. Production efficiency and consistency were characterized and the observed titer values of the viral harvest and the processed bulk were comparable to the expansion in embryonated eggs. The present study clearly establishes the suitability of MDCK cells as a substrate for the manufacture of a safe and viable LAIV.
Collapse
Affiliation(s)
- Milan Ganguly
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India.
| | - Leena Yeolekar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Parikshit Tyagi
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Umesh Sagar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Swapnil Narale
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | | | - Sham Tupe
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Kuntinath Wadkar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Nilesh Ingle
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Rajeev Dhere
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | | | | |
Collapse
|
17
|
Qiao Z, Yang D, Liu L, Liu Z, Wang J, He D, Wu H, Wang J, Ma Z. Genome-wide identification and characterization of long non-coding RNAs in MDCK cell lines with high and low tumorigenicities. Genomics 2020; 112:1077-1086. [DOI: 10.1016/j.ygeno.2019.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/03/2019] [Accepted: 08/06/2019] [Indexed: 10/26/2022]
|
18
|
Bissinger T, Fritsch J, Mihut A, Wu Y, Liu X, Genzel Y, Tan WS, Reichl U. Semi-perfusion cultures of suspension MDCK cells enable high cell concentrations and efficient influenza A virus production. Vaccine 2019; 37:7003-7010. [DOI: 10.1016/j.vaccine.2019.04.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/10/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
|
19
|
Cell-Based Quadrivalent Inactivated Influenza Virus Vaccine (Flucelvax ® Tetra/Flucelvax Quadrivalent ®): A Review in the Prevention of Influenza. Drugs 2019; 79:1337-1348. [PMID: 31372959 PMCID: PMC6874518 DOI: 10.1007/s40265-019-01176-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Manufacturing influenza virus vaccines using a mammalian cell line rather than embryonated chicken eggs may carry certain advantages. A quadrivalent inactivated influenza virus vaccine produced using the Madin Darby canine kidney cell line has been approved in the EU (Flucelvax® Tetra) and USA (Flucelvax Quadrivalent®; QIVc hereafter) for the prevention of influenza in adults and children. The clinical development of QIVc has built upon that of a cell-based trivalent influenza virus vaccine (TIVc) manufactured using the same processes; the additional influenza B strain contained in QIVc reduces the risk of the strain in the vaccine not matching that in circulation. Pivotal phase III clinical trials in adult and paediatric participants have demonstrated the immunogenicity of QIVc to be noninferior to that of TIVc formulations against shared strains and superior against the influenza B strain absent from each TIVc formulation. Protective efficacy data for TIVc is considered foundational for QIVc and, in a phase III clinical trial, TIVc was effective in protecting adults against antigenically matched influenza strains. Large real-world studies from the 2017/2018 US influenza season further support the prophylactic effectiveness of QIVc, with possible benefits over egg-based vaccines. QIVc was generally well tolerated in clinical trials. In adult and paediatric QIVc recipients, the most common solicited adverse reactions were injection site pain and headache. Reactogenicity was comparable to that of TIVc; no safety signals unique to QIVc emerged. Through circumventing concerns around egg adaptation, QIVc has the potential to be more effective than currently available egg-based quadrivalent vaccines.
Collapse
|
20
|
Trombetta CM, Marchi S, Manini I, Lazzeri G, Montomoli E. Challenges in the development of egg-independent vaccines for influenza. Expert Rev Vaccines 2019; 18:737-750. [DOI: 10.1080/14760584.2019.1639503] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giacomo Lazzeri
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
| |
Collapse
|
21
|
Barr IG, Donis RO, Katz JM, McCauley JW, Odagiri T, Trusheim H, Tsai TF, Wentworth DE. Cell culture-derived influenza vaccines in the severe 2017-2018 epidemic season: a step towards improved influenza vaccine effectiveness. NPJ Vaccines 2018; 3:44. [PMID: 30323955 PMCID: PMC6177469 DOI: 10.1038/s41541-018-0079-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 11/21/2022] Open
Abstract
The 2017–2018 seasonal influenza epidemics were severe in the US and Australia where the A(H3N2) subtype viruses predominated. Although circulating A(H3N2) viruses did not differ antigenically from that recommended by the WHO for vaccine production, overall interim vaccine effectiveness estimates were below historic averages (33%) for A(H3N2) viruses. The majority (US) or all (Australian) vaccine doses contained multiple amino-acid changes in the hemagglutinin protein, resulting from the necessary adaptation of the virus to embryonated hen’s eggs used for most vaccine manufacturing. Previous reports have suggested a potential negative impact of egg-driven substitutions on vaccine performance. With BARDA support, two vaccines licensed in the US are produced in cell culture: recombinant influenza vaccine (RIV, Flublok™) manufactured in insect cells and inactivated mammalian cell-grown vaccine (ccIIV, Flucelvax™). Quadrivalent ccIIV (ccIIV4) vaccine for the 2017–2018 influenza season was produced using an A(H3N2) seed virus propagated exclusively in cell culture and therefore lacking egg adaptative changes. Sufficient ccIIV doses were distributed (but not RIV doses) to enable preliminary estimates of its higher effectiveness relative to the traditional egg-based vaccines, with study details pending. The increased availability of comparative product-specific vaccine effectiveness estimates for cell-based and egg-based vaccines may provide critical clues to inform vaccine product improvements moving forward.
Collapse
Affiliation(s)
- Ian G Barr
- 1WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute For Infection And Immunity, 792 Elizabeth Street, Melbourne, 3000 Australia
| | - Ruben O Donis
- Biomedical Advanced Research and Development Authority, Influenza and Emerging Infectious Diseases Division, 300 Independence Avenue, SW, Washington, DC 20201 USA
| | - Jacqueline M Katz
- 3Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road MS A-20, Atlanta, GA 30329-4027 USA
| | - John W McCauley
- 4WHO Collaborating Centre for Reference and Research on Influenza, Crick Worldwide Influenza Centre, The Francis Crick Institute, 1, Midland Road, London, NW1 1AT UK
| | - Takato Odagiri
- WHO Collaborating Centre for Reference and Research on Influenza, National Institute of Infectious Diseases, Influenza Virus Research Center, 4-7-1 Gakuen, Musashi-Murayama-shi, Tokyo 208-0011 Japan
| | - Heidi Trusheim
- IDT Biologika GmbH, Am Pharmapark, 06861 Dessau-Rosslau, Germany
| | - Theodore F Tsai
- 7Takeda Vaccines (USA), 75 Sidney St, Cambridge, MA 02139 USA
| | - David E Wentworth
- 8Influenza Division, Centers for Disease Control and Prevention (CDC), 1600 Clifton Road MS A-20, Atlanta, GA 30329-4027 USA
| |
Collapse
|
22
|
Chen P, Zhang KH, Na T, Wang L, Yin WD, Yuan BZ, Wang JZ. The hUC-MSCs cell line CCRC-1 represents a novel, safe and high-yielding HDCs for the production of human viral vaccines. Sci Rep 2017; 7:12484. [PMID: 28970485 PMCID: PMC5624879 DOI: 10.1038/s41598-017-11997-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 08/31/2017] [Indexed: 12/20/2022] Open
Abstract
MRC-5 represents the most frequent human diploid cells (HDCs)-type cell substrate in the production of human viral vaccines. However, early-passage MRC-5 is diminishing and, due to both technical and ethical issues, it is extremely difficult to derive novel HDCs from fetal lung tissues, which are the common sources of HDCs. Our previous studies suggested that human umbilical cord may represent an alternative but convenient source of new HDCs. Here, we established a three-tiered cell banking system of a hUC-MSC line, designated previously as Cell Collection and Research Center-1 (CCRC-1). The full characterization indicated that the banked CCRC-1 cells were free from adventitious agents and remained non-tumorigenic. The CCRC-1 cells sustained its rapid proliferation even at passage 30 and were susceptible to the infection of a wide spectrum of viruses. Interestingly, the CCRC-1 cells showed much higher production of EV71 or Rubella viruses than MRC-5 and Vero cells when growing in serum-free medium. More importantly, the EV71 vaccine produced from CCRC-1 cells induced immunogenicity while eliciting no detectable toxicities in the tested mice. Collectively, these studies further supported that CCRC-1, and likely other hUC-MSCs as well, may serve as novel, safe and high-yielding HDCs for the production of human viral vaccines.
Collapse
Affiliation(s)
- Ping Chen
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, 100050, PR China.,Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, P.R. China
| | - Ke-Hua Zhang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, 100050, PR China
| | - Tao Na
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, 100050, PR China
| | - Lin Wang
- Sino Vac Biotech, Beijing, 100085, PR China
| | | | - Bao-Zhu Yuan
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, 100050, PR China.
| | - Jun-Zhi Wang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, 100050, PR China.
| |
Collapse
|
23
|
Manini I, Trombetta CM, Lazzeri G, Pozzi T, Rossi S, Montomoli E. Egg-Independent Influenza Vaccines and Vaccine Candidates. Vaccines (Basel) 2017; 5:E18. [PMID: 28718786 PMCID: PMC5620549 DOI: 10.3390/vaccines5030018] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 11/16/2022] Open
Abstract
Vaccination remains the principal way to control seasonal infections and is the most effective method of reducing influenza-associated morbidity and mortality. Since the 1940s, the main method of producing influenza vaccines has been an egg-based production process. However, in the event of a pandemic, this method has a significant limitation, as the time lag from strain isolation to final dose formulation and validation is six months. Indeed, production in eggs is a relatively slow process and production yields are both unpredictable and highly variable from strain to strain. In particular, if the next influenza pandemic were to arise from an avian influenza virus, and thus reduce the egg-laying hen population, there would be a shortage of embryonated eggs available for vaccine manufacturing. Although the production of egg-derived vaccines will continue, new technological developments have generated a cell-culture-based influenza vaccine and other more recent platforms, such as synthetic influenza vaccines.
Collapse
Affiliation(s)
- Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Claudia Maria Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Giacomo Lazzeri
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Teresa Pozzi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Stefania Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
- VisMederi S.r.l., Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy.
| |
Collapse
|
24
|
Diez-Domingo J, de Martino M, Lopez JGS, Zuccotti GV, Icardi G, Villani A, Moreno-Perez D, Hernández MM, Aldeán JÁ, Mateen AA, Enweonye I, de Rooij R, Chandra R. Safety and tolerability of cell culture-derived and egg-derived trivalent influenza vaccines in 3 to <18-year-old children and adolescents at risk of influenza-related complications. Int J Infect Dis 2016; 49:171-8. [PMID: 27343983 DOI: 10.1016/j.ijid.2016.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/15/2016] [Accepted: 06/16/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND This descriptive, non-comparative, phase III study evaluated the safety and tolerability of cell culture-derived (TIVc) and egg-derived (TIV) seasonal influenza vaccines in children at risk of influenza-related complications. METHODS Four hundred and thirty subjects were randomized 2:1 to TIVc or TIV. Subjects aged 3 to <9 years received one dose (if previously vaccinated, n=89) or two doses (if not previously vaccinated, n=124) of the study vaccines; the 9 to <18-year-olds (n=213) received one dose. Reactogenicity was assessed for 7 days after vaccination; safety was monitored for 6 months. RESULTS After any vaccination, the most frequently reported solicited local adverse event (AE) was tenderness/pain (TIVc 44%, 66%, 53% and TIV 56%, 51%, 65% in the age groups 3 to <6 years, 6 to <9 years, and 9 to <18 years, respectively) and the systemic AE was irritability (22% TIVc, 24% TIV) in 3 to <6-year-olds and headache in 6 to <9-year-olds (20% TIVc, 13% TIV) and 9 to <18-year-olds (21% TIVc, 26% TIV). There were no cases of severe fever (≥40°C). No vaccine-related serious AEs were noted. New onset of chronic disease was reported in ≤1% of subjects. CONCLUSION TIVc and TIV had acceptable tolerability and similar safety profiles in at-risk children (NCT01998477).
Collapse
Affiliation(s)
| | | | | | - Gian Vincenzo Zuccotti
- Department of Paediatrics, Children Hospital V. Buzzi, University of Milan, Milan, Italy
| | - Giancarlo Icardi
- Department of Health Sciences, University of Genoa and I.R.C.C.S. University Hospital, San Martino-IST National Institute for Cancer Research, Genoa, Italy
| | - Alberto Villani
- Department of General Paediatrics and Infectious Diseases, Bambino Gesù Children Hospital, Rome, Italy
| | | | | | | | - Ahmed Abdul Mateen
- Novartis Pharmaceuticals Canada Inc., 385, Bouchard Blvd, Dorval, Quebec H9S 1A9, Canada.
| | | | | | - Richa Chandra
- Novartis Vaccines and Diagnostics Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
25
|
André M, Reghin S, Boussard E, Lempereur L, Maisonneuve S. Universal real-time PCR assay for quantitation and size evaluation of residual cell DNA in human viral vaccines. Biologicals 2016; 44:139-49. [DOI: 10.1016/j.biologicals.2016.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/26/2015] [Accepted: 03/05/2016] [Indexed: 12/15/2022] Open
|
26
|
Trombetta CM, Montomoli E. Influenza immunology evaluation and correlates of protection: a focus on vaccines. Expert Rev Vaccines 2016; 15:967-76. [PMID: 26954563 DOI: 10.1586/14760584.2016.1164046] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Vaccination is the most effective method of controlling seasonal influenza infections and preventing possible pandemic events. Although influenza vaccines have been licensed and used for decades, the potential correlates of protection induced by these vaccines are still a matter of discussion. Currently, inactivated vaccines are the most common and the haemagglutination inhibition antibody titer is regarded as an immunological correlate of protection and the best available parameter for predicting protection from influenza infection. However, the assay shows some limitations, such as its low sensitivity to B and avian strains and inter-laboratory variability. Additional assays and next-generation vaccines have been evaluated to overcome the limitations of the traditional serological techniques and to elicit broad immune responses, underlining the need to revise the current correlates of protection. The aim of this review is to provide an overview of the current scenario regarding the immunological evaluation and correlates of protection of influenza vaccines.
Collapse
Affiliation(s)
| | - Emanuele Montomoli
- a Department of Molecular and Developmental Medicine , University of Siena , Siena , Italy.,b VisMederi srl , Enterprise of services in Life Sciences , Siena , Italy
| |
Collapse
|
27
|
Hegde NR. Cell culture-based influenza vaccines: A necessary and indispensable investment for the future. Hum Vaccin Immunother 2016; 11:1223-34. [PMID: 25875691 DOI: 10.1080/21645515.2015.1016666] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The traditional platform of using embryonated chicken eggs for the production of influenza vaccines has several drawbacks including the inability to meet the volume of required doses in the case of widespread epidemics and pandemics. Cell culture platforms have therefore been explored in the last 2 decades, and have attracted further attention following the H1N1 pandemic outbreak. This platform, while not the most economical for large-scale production, has several advantages, and can supplement the vaccine requirement when needed. Recent developments in production technologies have contributed greatly to fine-tuning this platform. In combination with other technologies such as live attenuated and recombinant protein or virus-like particle vaccines, and different adjuvants and delivery systems, cell culture-based influenza vaccine platform can be used both for production of seasonal vaccine, and to mitigate vaccine shortages in pandemic situations.
Collapse
Affiliation(s)
- Nagendra R Hegde
- a Ella Foundation; Genome Valley; Turkapally , Shameerpet Mandal , Hyderabad , India
| |
Collapse
|
28
|
Aubrit F, Perugi F, Léon A, Guéhenneux F, Champion-Arnaud P, Lahmar M, Schwamborn K. Cell substrates for the production of viral vaccines. Vaccine 2015; 33:5905-12. [PMID: 26187258 DOI: 10.1016/j.vaccine.2015.06.110] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/12/2015] [Accepted: 06/26/2015] [Indexed: 11/20/2022]
Abstract
Vaccines have been used for centuries to protect people and animals against infectious diseases. For vaccine production, it has become evident that cell culture technology can be considered as a key milestone and has been the result of decades of progress. The development and implementation of cell substrates have permitted massive and safe production of viral vaccines. The demand in new vaccines against emerging viral diseases, the increasing vaccine production volumes, and the stringent safety rules for manufacturing have made cell substrates mandatory viral vaccine producer factories. In this review, we focus on cell substrates for the production of vaccines against human viral diseases. Depending on the nature of the vaccine, choice of the cell substrate is critical. Each manufacturer intending to develop a new vaccine candidate should assess several cell substrates during the early development phase in order to select the most convenient for the application. First, as vaccine safety is quite naturally a central concern of Regulatory Agencies, the cell substrate has to answer the regulatory rules stringency. In addition, the cell substrate has to be competitive in terms of viral-specific production yields and manufacturing costs. No cell substrate, even the so-called "designer" cell lines, is able to fulfil all the requested criteria for all viral vaccines. Therefore, the availability of a variety of cell substrates for vaccine production is essential because it improves the chance to successfully respond to the current and future needs of vaccines linked to new emerging or re-emerging infectious diseases (e.g. pandemic flu, Ebola, and Chikungunya outbreaks).
Collapse
Affiliation(s)
- Françoise Aubrit
- Vaccines Research & Discovery Department, Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| | - Fabien Perugi
- Vaccines Research & Discovery Department, Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| | - Arnaud Léon
- Vaccines Research & Discovery Department, Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| | - Fabienne Guéhenneux
- Vaccines Research & Discovery Department, Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| | - Patrick Champion-Arnaud
- Vaccines Research & Discovery Department, Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| | - Mehdi Lahmar
- Vaccines Research & Discovery Department, Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| | - Klaus Schwamborn
- Vaccines Research & Discovery Department, Valneva SE, 6 rue Alain Bombard, 44800 Saint-Herblain, France.
| |
Collapse
|
29
|
Production of canine adenovirus type 2 in serum-free suspension cultures of MDCK cells. Appl Microbiol Biotechnol 2015; 99:7059-68. [DOI: 10.1007/s00253-015-6636-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/19/2015] [Accepted: 04/22/2015] [Indexed: 10/23/2022]
|
30
|
Choi EH, Song MS, Park SJ, Pascua PNQ, Baek YH, Kwon HI, Kim EH, Kim S, Jang HK, Poo H, Kim CJ, Choi YK. Development of a dual-protective live attenuated vaccine against H5N1 and H9N2 avian influenza viruses by modifying the NS1 gene. Arch Virol 2015; 160:1729-40. [PMID: 25959557 DOI: 10.1007/s00705-015-2442-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/26/2015] [Indexed: 11/26/2022]
Abstract
An increasing number of outbreaks of avian influenza H5N1 and H9N2 viruses in poultry have caused serious economic losses and raised concerns for human health due to the risk of zoonotic transmission. However, licensed H5N1 and H9N2 vaccines for animals and humans have not been developed. Thus, to develop a dual H5N1 and H9N2 live-attenuated influenza vaccine (LAIV), the HA and NA genes from a virulent mouse-adapted avian H5N2 (A/WB/Korea/ma81/06) virus and a recently isolated chicken H9N2 (A/CK/Korea/116/06) virus, respectively, were introduced into the A/Puerto Rico/8/34 backbone expressing truncated NS1 proteins (NS1-73, NS1-86, NS1-101, NS1-122) but still possessing a full-length NS gene. Two H5N2/NS1-LAIV viruses (H5N2/NS1-86 and H5N2/NS1-101) were highly attenuated compared with the full-length and remaining H5N2/NS-LAIV viruses in a mouse model. Furthermore, viruses containing NS1 modifications were found to induce more IFN-β activation than viruses with full-length NS1 proteins and were correspondingly attenuated in mice. Intranasal vaccination with a single dose (10(4.0) PFU/ml) of these viruses completely protected mice from a lethal challenge with the homologous A/WB/Korea/ma81/06 (H5N2), heterologous highly pathogenic A/EM/Korea/W149/06 (H5N1), and heterosubtypic highly virulent mouse-adapted H9N2 viruses. This study clearly demonstrates that the modified H5N2/NS1-LAIV viruses attenuated through the introduction of mutations in the NS1 coding region display characteristics that are desirable for live attenuated vaccines and hold potential as vaccine candidates for mammalian hosts.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Chickens
- Female
- Humans
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H9N2 Subtype/genetics
- Influenza A Virus, H9N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Poultry Diseases/immunology
- Poultry Diseases/prevention & control
- Poultry Diseases/virology
- Vaccination
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Viral Nonstructural Proteins/administration & dosage
- Viral Nonstructural Proteins/genetics
- Viral Nonstructural Proteins/immunology
Collapse
Affiliation(s)
- Eun-hye Choi
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, 12 Gaeshin-Dong Heungduk-Ku, Cheongju, 361-763, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Manini I, Domnich A, Amicizia D, Rossi S, Pozzi T, Gasparini R, Panatto D, Montomoli E. Flucelvax (Optaflu) for seasonal influenza. Expert Rev Vaccines 2015; 14:789-804. [DOI: 10.1586/14760584.2015.1039520] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
32
|
Omeir R, Thomas R, Teferedegne B, Williams C, Foseh G, Macauley J, Brinster L, Beren J, Peden K, Breen M, Lewis AM. A novel canine kidney cell line model for the evaluation of neoplastic development: karyotype evolution associated with spontaneous immortalization and tumorigenicity. Chromosome Res 2015; 23:663-80. [PMID: 25957863 PMCID: PMC4666904 DOI: 10.1007/s10577-015-9474-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/12/2015] [Accepted: 04/14/2015] [Indexed: 01/01/2023]
Abstract
The molecular mechanisms underlying spontaneous neoplastic transformation in cultured mammalian cells remain poorly understood, confounding recognition of parallels with the biology of naturally occurring cancer. The broad use of tumorigenic canine cell lines as research tools, coupled with the accumulation of cytogenomic data from naturally occurring canine cancers, makes the domestic dog an ideal system in which to investigate these relationships. We developed a canine kidney cell line, CKB1-3T7, which allows prospective examination of the onset of spontaneous immortalization and tumorigenicity. We documented the accumulation of cytogenomic aberrations in CKB1-3T7 over 24 months in continuous culture. The majority of aberrations emerged in parallel with key phenotypic changes in cell morphology, growth kinetics, and tumor incidence and latency. Focal deletion of CDKN2A/B emerged first, preceding the onset and progression of tumorigenic potential, and progressed to a homozygous deletion across the cell population during extended culture. Interestingly, CKB1-3T7 demonstrated a tumorigenic phenotype in vivo prior to exhibiting loss of contact inhibition in vitro. We also performed the first genome-wide characterization of the canine tumorigenic cell line MDCK, which also exhibited CDKN2A/B deletion. MDCK and CKB1-3T7 cells shared several additional aberrations that we have reported previously as being highly recurrent in spontaneous canine cancers, many of which, as with CDKN2A/B deletion, are evolutionarily conserved in their human counterparts. The conservation of these molecular events across multiple species, in vitro and in vivo, despite their contrasting karyotypic architecture, is a powerful indicator of a common mechanism underlying emerging neoplastic activity. Through integrated cytogenomic and phenotypic characterization of serial passages of CKB1-3T7 from initiation to development of a tumorigenic phenotype, we present a robust and readily accessible model (to be made available through the American Type Culture Collection) of spontaneous neoplastic transformation that overcomes many of the limitations of earlier studies.
Collapse
Affiliation(s)
- R Omeir
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - R Thomas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA.,Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, 27607, USA
| | - B Teferedegne
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - C Williams
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA
| | - G Foseh
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - J Macauley
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - L Brinster
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, 20892, USA
| | - J Beren
- Office of Counter-Terrorism and Emergency Coordination, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - K Peden
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA
| | - M Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC, 27607, USA. .,Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, NC, 27607, USA. .,Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 27599, USA. .,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27607, USA.
| | - A M Lewis
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| |
Collapse
|
33
|
Donis RO, Chen IM, Davis CT, Foust A, Hossain MJ, Johnson A, Klimov A, Loughlin R, Xu X, Tsai T, Blayer S, Trusheim H, Colegate T, Fox J, Taylor B, Hussain A, Barr I, Baas C, Louwerens J, Geuns E, Lee MS, Venhuizen O, Neumeier E, Ziegler T. Performance characteristics of qualified cell lines for isolation and propagation of influenza viruses for vaccine manufacturing. Vaccine 2014; 32:6583-90. [PMID: 24975811 PMCID: PMC5915289 DOI: 10.1016/j.vaccine.2014.06.045] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/22/2014] [Accepted: 06/11/2014] [Indexed: 01/18/2023]
Abstract
Cell culture is now available as a method for the production of influenza vaccines in addition to eggs. In accordance with currently accepted practice, viruses recommended as candidates for vaccine manufacture are isolated and propagated exclusively in hens' eggs prior to distribution to manufacturers. Candidate vaccine viruses isolated in cell culture are not available to support vaccine manufacturing in mammalian cell bioreactors so egg-derived viruses have to be used. Recently influenza A (H3N2) viruses have been difficult to isolate directly in eggs. As mitigation against this difficulty, and the possibility of no suitable egg-isolated candidate viruses being available, it is proposed to consider using mammalian cell lines for primary isolation of influenza viruses as candidates for vaccine production in egg and cell platforms. To investigate this possibility, we tested the antigenic stability of viruses isolated and propagated in cell lines qualified for influenza vaccine manufacture and subsequently investigated antigen yields of such viruses in these cell lines at pilot-scale. Twenty influenza A and B-positive, original clinical specimens were inoculated in three MDCK cell lines. The antigenicity of recovered viruses was tested by hemagglutination inhibition using ferret sera against contemporary vaccine viruses and the amino acid sequences of the hemagglutinin and neuraminidase were determined. MDCK cell lines proved to be highly sensitive for virus isolation. Compared to the virus sequenced from the original specimen, viruses passaged three times in the MDCK lines showed up to 2 amino acid changes in the hemagglutinin. Antigenic stability was also established by hemagglutination inhibition titers comparable to those of the corresponding reference virus. Viruses isolated in any of the three MDCK lines grew reasonably well but variably in three MDCK cells and in VERO cells at pilot-scale. These results indicate that influenza viruses isolated in vaccine certified cell lines may well qualify for use in vaccine production.
Collapse
Affiliation(s)
- Ruben O. Donis
- Corresponding author Influenza Division, Centers for Diseases Control and Prevention (CDC), National Center for Immunization and Respiratory Diseases, Influenza Division, 1600 Clifton Road, Mailstop A-20, Atlanta, GA 30333, USA. Tel.: +1 404 639 4968; fax: +1 404 639 2350. ,
| | | | - i-Mei Chen
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - C Todd Davis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Angie Foust
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - M. Jaber Hossain
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Adam Johnson
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Alexander Klimov
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
- National Institute for Health and Welfare (THL), Helsinki, Finland, Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA USA
| | - Rosette Loughlin
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Xiyan Xu
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Theodore Tsai
- Novartis Vaccines and Diagnostics, Cambridge, MA, USA
| | - Simone Blayer
- Novartis Vaccines and Diagnostics, GmbH & Co. KG, Marburg, Germany
| | - Heidi Trusheim
- Novartis Vaccines and Diagnostics, GmbH & Co. KG, Marburg, Germany
| | | | - John Fox
- Novartis Vaccines and Diagnostics, Liverpool, UK
| | | | | | - Ian Barr
- WHO Collaborating Centre for Reference and Research on Influenza, North Melbourne, Victoria, Australia
| | - Chantal Baas
- WHO Collaborating Centre for Reference and Research on Influenza, North Melbourne, Victoria, Australia
| | | | - Ed Geuns
- Abbott Bioiogicais B.V., Weesp, The Netherlands
| | | | | | | | - Thedi Ziegler
- National Institute for Health and Welfare (THL), Helsinki, Finland, Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA USA
| |
Collapse
|
34
|
Trombetta CM, Perini D, Mather S, Temperton N, Montomoli E. Overview of Serological Techniques for Influenza Vaccine Evaluation: Past, Present and Future. Vaccines (Basel) 2014; 2:707-34. [PMID: 26344888 PMCID: PMC4494249 DOI: 10.3390/vaccines2040707] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/25/2014] [Accepted: 09/22/2014] [Indexed: 12/12/2022] Open
Abstract
Serological techniques commonly used to quantify influenza-specific antibodies include the Haemagglutination Inhibition (HI), Single Radial Haemolysis (SRH) and Virus Neutralization (VN) assays. HI and SRH are established and reproducible techniques, whereas VN is more demanding. Every new influenza vaccine needs to fulfil the strict criteria issued by the European Medicines Agency (EMA) in order to be licensed. These criteria currently apply exclusively to SRH and HI assays and refer to two different target groups-healthy adults and the elderly, but other vaccine recipient age groups have not been considered (i.e., children). The purpose of this timely review is to highlight the current scenario on correlates of protection concerning influenza vaccines and underline the need to revise the criteria and assays currently in use. In addition to SRH and HI assays, the technical advantages provided by other techniques such as the VN assay, pseudotype-based neutralization assay, neuraminidase and cell-mediated immunity assays need to be considered and regulated via EMA criteria, considering the many significant advantages that they could offer for the development of effective vaccines.
Collapse
Affiliation(s)
- Claudia Maria Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 53100 Siena, Italy.
| | - Daniele Perini
- VisMederi srl, Enterprise in Life Sciences, Via Fiorentina 1, 53100 Siena, Italy.
| | - Stuart Mather
- Viral Pseudotype Unit, School of Pharmacy, University of Kent, Chatham Maritime, Kent ME4 4TB, UK.
| | - Nigel Temperton
- Viral Pseudotype Unit, School of Pharmacy, University of Kent, Chatham Maritime, Kent ME4 4TB, UK.
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 53100 Siena, Italy.
- VisMederi srl, Enterprise in Life Sciences, Via Fiorentina 1, 53100 Siena, Italy.
| |
Collapse
|
35
|
Huber VC. Influenza vaccines: from whole virus preparations to recombinant protein technology. Expert Rev Vaccines 2014; 13:31-42. [PMID: 24192014 DOI: 10.1586/14760584.2014.852476] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Vaccination against influenza represents our most effective form of prevention. Historical approaches toward vaccine creation and production have yielded highly effective vaccines that are safe and immunogenic. Despite their effectiveness, these historical approaches do not allow for the incorporation of changes into the vaccine in a timely manner. In 2013, a recombinant protein-based vaccine that induces immunity toward the influenza virus hemagglutinin was approved for use in the USA. This vaccine represents the first approved vaccine formulation that does not require an influenza virus intermediate for production. This review presents a brief history of influenza vaccines, with insight into the potential future application of vaccines generated using recombinant technology.
Collapse
Affiliation(s)
- Victor C Huber
- Division of Basic Biomedical Sciences, University of South Dakota, 414 E Clark Street, Vermillion, SD 57069, USA
| |
Collapse
|
36
|
Development and preclinical testing of HNVAC, a cell culture-based H1N1 pandemic influenza vaccine from India. Vaccine 2014; 32:3636-43. [DOI: 10.1016/j.vaccine.2014.04.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 12/24/2022]
|
37
|
Dormitzer PR. Rapid Production of Synthetic Influenza Vaccines. Curr Top Microbiol Immunol 2014; 386:237-73. [DOI: 10.1007/82_2014_399] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
38
|
Lugovtsev VY, Melnyk D, Weir JP. Heterogeneity of the MDCK cell line and its applicability for influenza virus research. PLoS One 2013; 8:e75014. [PMID: 24058646 PMCID: PMC3772841 DOI: 10.1371/journal.pone.0075014] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/08/2013] [Indexed: 11/18/2022] Open
Abstract
Single-cell clones have been established from the MDCK cell line, characterized for their morphology and evaluated for their suitability for influenza virus research. Three discrete cell morphotypes were identified using light microscopy. Besides morphological features, the cell types can be distinguished by the level of expression of surface glycans recognized by peanut agglutinin (PNA). All clones were susceptible to infection by influenza viruses of different subtypes of influenza A virus (H1N1, H1N1pdm09, H3N2, H5N1) and influenza B virus, and all possessed on their surface terminally sialylated glycans with both types of glycosidic linkage (α2-3 and α2-6). The Type-1 cell lines were able to support a multicycle replication of influenza A and B viruses without help of an exogenous trypsin. In contrast, cell lines exhibiting Type-2 morphology were unable to support multicycle replication of influenza A viruses without trypsin supplementation. Western blot analysis of the hemagglutinin of H1N1 strains demonstrated that Type-2 cells were deficient in production of proteolytically activated hemagglutinin (no cleavage between HA1/HA2 was observed). HA1/HA2 cleavage of influenza B viruses in the Type-2 cells was also significantly impaired, but not completely abrogated, producing sufficient amount of activated HA to support efficient virus replication without trypsin. In contrast, all clones of Type-1 cells were able to produce proteolytically activated hemagglutinin of influenza A and B viruses. However, the growth kinetics and plaque size of influenza A viruses varied significantly in different clones. Influenza B virus also showed different plaque size, with the biggest plaque formation in the Type-2 cells, although the growth kinetics and peak infectivity titers were similar in all clones. Taken together, the study demonstrates that the population of original MDCK cells is represented by various types of cells that differ in their capacities to support replication of influenza A and B viruses.
Collapse
Affiliation(s)
- Vladimir Y. Lugovtsev
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
- * E-mail:
| | - Darya Melnyk
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Jerry P. Weir
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| |
Collapse
|
39
|
Loebermann M, Voss U, Meyer S, Bosse D, Fritzsche C, Klammt S, Frimmel S, Riebold D, Reisinger EC. Clinical trial to evaluate the safety and immunogenicity of a trivalent surface antigen seasonal influenza vaccine produced in mammalian cell culture and administered to young and elderly adults with and without A(H1N1) pre-vaccination. PLoS One 2013; 8:e70866. [PMID: 23976960 PMCID: PMC3745456 DOI: 10.1371/journal.pone.0070866] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 06/19/2013] [Indexed: 11/27/2022] Open
Abstract
UNLABELLED Vaccination against influenza is an important means of reducing morbidity and mortality in subjects at risk. The prevalent viral strains responsible for seasonal epidemics usually change annually, but the WHO recommendations for the 2011/2012-season in the Northern hemisphere included the same antigens as for the previous season. We conducted a single-center, single-arm study involving 62 younger (18-60 years) and 64 older (>60 years) adults to test the immunogenicity, safety and tolerability of a trivalent surface antigen, inactivated influenza vaccine produced in mammalian cell-culture. The vaccine contained 15 µg hemagglutinin of each of the virus strains recommended for the 2011-2012 Northern hemisphere winter season (A/California/7/09 (H1N1)-; A/Perth/16/09 (H3N2)-; B/Brisbane/60/08-like strain) in a non-adjuvanted preservative-free formulation. Antibody response was measured by hemagglutination inhibition 21 days after immunization. Adverse events and safety were assessed using subject diary cards and telephone interviews. Seroconversion or a 4-fold antibody increase in antibody titers was detectable against A(H1N1) in 68% of both younger and older adults, against A(H3N2) in 53% and 27%, and against the B influenza strain in 35% and 17%. Antibody titers of 40 or more were observed against A(H1N1) in 87% and 90% of younger and older adults, against A(H3N2) in 98% and 98%, and against the B influenza strain in 93% and 90%. Pre-vaccination antibody titers were protective against A(H1N1), A(H3N2) and B in 38%, 58% and 58%, respectively, of younger and in 43%, 88% and 70% of older adults. Among subjects with previous A(H1N1) vaccination only 48% of younger and 47% of older adults had protective A(H1N1) antibodies at inclusion. Adverse reactions were generally mild. The most frequently reported reactions were pain at the injection site, myalgia and fatigue. The vaccine generated protective antibodies against all three viral strains and had an acceptable safety profile in both younger and older adults. TRIAL REGISTRATION ClinicalTrials.govNCT01422512.
Collapse
Affiliation(s)
- Micha Loebermann
- Department of Tropical Medicine, Infectious Diseases and Nephrology, University of Rostock, Rostock, Germany
| | - Ulrich Voss
- Department of Tropical Medicine, Infectious Diseases and Nephrology, University of Rostock, Rostock, Germany
| | - Seetha Meyer
- Novartis Vaccines and Diagnostics GmbH, Marburg, Germany
| | - Dietrich Bosse
- Novartis Vaccines and Diagnostics GmbH, Marburg, Germany
| | - Carlos Fritzsche
- Department of Tropical Medicine, Infectious Diseases and Nephrology, University of Rostock, Rostock, Germany
| | - Sebastian Klammt
- Department of Tropical Medicine, Infectious Diseases and Nephrology, University of Rostock, Rostock, Germany
| | - Silvius Frimmel
- Department of Tropical Medicine, Infectious Diseases and Nephrology, University of Rostock, Rostock, Germany
| | - Diana Riebold
- Department of Tropical Medicine, Infectious Diseases and Nephrology, University of Rostock, Rostock, Germany
| | - Emil C. Reisinger
- Department of Tropical Medicine, Infectious Diseases and Nephrology, University of Rostock, Rostock, Germany
| |
Collapse
|
40
|
An MDCK cell culture-derived formalin-inactivated influenza virus whole-virion vaccine from an influenza virus library confers cross-protective immunity by intranasal administration in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:998-1007. [PMID: 23637045 DOI: 10.1128/cvi.00024-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
It is currently impossible to predict the next pandemic influenza virus strain. We have thus established a library of influenza viruses of all hemagglutinin and neuraminidase subtypes and their genes. In this article, we examine the applicability of a rapid production model for the preparation of vaccines against emerging pandemic influenza viruses. This procedure utilizes the influenza virus library, cell culture-based vaccine production, and intranasal administration to induce a cross-protective immune response. First, an influenza virus reassortant from the library, A/duck/Hokkaido/Vac-3/2007 (H5N1), was passaged 22 times (P22) in Madin-Darby canine kidney (MDCK) cells. The P22 virus had a titer of >2 ×10(8) PFU/ml, which was 40 times that of the original strain, with 4 point mutations, which altered amino acids in the deduced protein sequences encoded by the PB2 and PA genes. We then produced a formalin-inactivated whole-virion vaccine from the MDCK cell-cultured A/duck/Hokkaido/Vac-3/2007 (H5N1) P22 virus. Intranasal immunization of mice with this vaccine protected them against challenges with lethal influenza viruses of homologous and heterologous subtypes. We further demonstrated that intranasal immunization with the vaccine induced cross-reactive neutralizing antibody responses against the homotypic H5N1 influenza virus and its antigenic variants and cross-reactive cell-mediated immune responses to the homologous virus, its variants within a subtype, and even an influenza virus of a different subtype. These results indicate that a rapid model for emergency vaccine production may be effective for producing the next generation of pandemic influenza virus vaccines.
Collapse
|
41
|
Assessment of the immunogenicity and safety of varying doses of an MF59®-adjuvanted cell culture-derived A/H1N1 pandemic influenza vaccine in Japanese paediatric, adult and elderly subjects. Vaccine 2012; 30:5030-7. [PMID: 22472791 DOI: 10.1016/j.vaccine.2012.03.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 03/10/2012] [Accepted: 03/21/2012] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Effective vaccination strategies are required to combat future influenza pandemics. Here we report the results of three independent clinical trials performed in Japan to assess the immunogenicity, tolerability and safety of varying doses of a cell culture-derived MF59(®)-adjuvanted A/H1N1 pandemic vaccine in healthy Japanese paediatric, adult and elderly subjects. METHODS One hundred and twenty-three children (6 months-18 years), and 200 adults (19-60 years) were randomly assigned in a 1:1 ratio to receive two doses of vaccine containing either 7.5 μg antigen with a full (9.75 mg) adjuvant dose, or 3.75 μg antigen with a half (4.875 mg) adjuvant dose. One hundred elderly (≥ 61 years) subjects received only the low antigen/adjuvant vaccine formulation. Immunogenicity was assessed by haemagglutination inhibition assay at baseline and three weeks after the first and second vaccine doses on Days 22 and 43, respectively. Solicited and unsolicited adverse reactions were recorded for seven and 21 days post-immunization, respectively. RESULTS In adult and elderly subjects, a single low antigen/adjuvant dose vaccination was sufficient to meet all of the three European licensure criteria established for influenza vaccines. One high, or two low antigen/adjuvant dose vaccinations were required to meet the licensure criteria in paediatric subjects. Both vaccine formulations were well tolerated, with the majority of adverse reactions mild to moderate in severity. None of the five serious adverse events reported throughout the three trials were considered to be vaccine-related by the investigators. CONCLUSION The use of MF59 adjuvant allows for much reduced vaccine antigen content, and a single dose administration schedule in adults and the elderly. The production of pandemic vaccine using modern cell culture techniques is highly advantageous in terms of the quantity, quality, and rapidity of antigen production; these benefits, in combination with the use of MF59, maximize manufacturing capacity and global vaccine supply. These data support the suitability of the investigational vaccine for use in the Japanese paediatric, adult, and elderly populations.
Collapse
|
42
|
New strategies for the development of H5N1 subtype influenza vaccines: progress and challenges. BioDrugs 2012; 25:285-98. [PMID: 21942913 DOI: 10.1007/bf03256169] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The emergence and spread of highly pathogenic avian influenza (H5N1) viruses among poultry in Asia, the Middle East, and Africa have fueled concerns of a possible human pandemic, and spurred efforts towards developing vaccines against H5N1 influenza viruses, as well as improving vaccine production methods. In recent years, promising experimental reverse genetics-derived H5N1 live attenuated vaccines have been generated and characterized, including vaccines that are attenuated through temperature-sensitive mutation, modulation of the interferon antagonist protein, or disruption of the M2 protein. Live attenuated influenza virus vaccines based on each of these modalities have conferred protection against homologous and heterologous challenge in animal models of influenza virus infection. Alternative vaccine strategies that do not require the use of live virus, such as virus-like particle (VLP) and DNA-based vaccines, have also been vigorously pursued in recent years. Studies have demonstrated that influenza VLP vaccination can confer homologous and heterologous protection from lethal challenge in a mouse model of infection. There have also been improvements in the formulation and production of vaccines following concerns over the threat of H5N1 influenza viruses. The use of novel substrates for the growth of vaccine virus stocks has been intensively researched in recent years, and several candidate cell culture-based systems for vaccine amplification have emerged, including production systems based on Madin-Darby canine kidney, Vero, and PerC6 cell lines. Such systems promise increased scalability of product, and reduced reliance on embryonated chicken eggs as a growth substrate. Studies into the use of adjuvants have shown that oil-in-water-based adjuvants can improve the immunogenicity of inactivated influenza vaccines and conserve antigen in such formulations. Finally, efforts to develop more broadly cross-protective immunization strategies through the inclusion of conserved influenza virus antigens in vaccines have led to experimental vaccines based on the influenza hemagglutinin (HA) stem domain. Such vaccines have been shown to confer protection from lethal challenge in mouse models of influenza virus infection. Through further development, vaccines based on the HA stem have the potential to protect vaccinated individuals against unanticipated pandemic and epidemic influenza virus strains. Overall, recent advances in experimental vaccines and in vaccine production processes provide the potential to lower mortality and morbidity resulting from influenza infection.
Collapse
|
43
|
Eggers M, Roth B, Schweiger B, Schmid M, Gregersen JP, Enders M. Comparison of the novel ResPlex III assay and existing techniques for the detection and subtyping of influenza virus during the influenza season 2006-2007. Eur J Clin Microbiol Infect Dis 2011; 31:1257-65. [PMID: 22012658 PMCID: PMC3346937 DOI: 10.1007/s10096-011-1437-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 09/22/2011] [Indexed: 11/15/2022]
Abstract
Influenza virus is a major cause of disease worldwide. The accurate detection and further subtyping of influenza A viruses are important for epidemiologic surveillance, and subsequent comprehensive characterization of circulating influenza viruses is essential for the selection of an optimal vaccine composition. ResPlex III is a new multiplex reverse transcriptase polymerase chain reaction (RT-PCR)-based method for detecting, typing, and subtyping influenza virus in clinical specimens. The ResPlex III assay was compared with other methods with respect to sensitivity and accuracy, using 450 clinical specimens obtained from subjects throughout Germany during the 2006–2007 influenza season. Samples were analyzed for the presence of influenza virus in Madin-Darby canine kidney (MDCK) cells by rapid cell culture using peroxidase staining and conventional cell culture confirmed by hemagglutination inhibition assay, a rapid diagnostic assay (Directigen Flu A+B test; BD Diagnostic Systems, Heidelberg, Germany), in-house real-time RT-PCR (RRT-PCR), and ResPlex III (Qiagen, Hilden, Germany). ResPlex III had the highest sensitivity for detecting influenza virus in clinical specimens, followed by in-house RRT-PCR (96% compared with ResPlex III). Conventional cell culture in MDCK cells, rapid culture, and quick test assays were substantially less sensitive (55%, 72%, and 39%, respectively). Virus subtyping results were identical using ResPlex III and the standard virological subtyping method, hemagglutination inhibition. ResPlex III is a quick, accurate, and sensitive assay for detecting and typing influenza A and B viruses and subtyping influenza A viruses in clinical specimens, and might be considered for a supplemental role in worldwide seasonal and pandemic influenza surveillance.
Collapse
Affiliation(s)
- M Eggers
- Laboratory Prof. G. Enders and Partner & Institute of Virology, Infectious Diseases and Epidemiology e.V., Rosenbergstrasse 85, 70193, Stuttgart, Germany.
| | | | | | | | | | | |
Collapse
|
44
|
Petiot E, Jacob D, Lanthier S, Lohr V, Ansorge S, Kamen AA. Metabolic and kinetic analyses of influenza production in perfusion HEK293 cell culture. BMC Biotechnol 2011; 11:84. [PMID: 21884612 PMCID: PMC3175177 DOI: 10.1186/1472-6750-11-84] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 09/01/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell culture-based production of influenza vaccine remains an attractive alternative to egg-based production. Short response time and high production yields are the key success factors for the broader adoption of cell culture technology for industrial manufacturing of pandemic and seasonal influenza vaccines. Recently, HEK293SF cells have been successfully used to produce influenza viruses, achieving hemagglutinin (HA) and infectious viral particle (IVP) titers in the highest ranges reported to date. In the same study, it was suggested that beyond 4 × 10(6) cells/mL, viral production was limited by a lack of nutrients or an accumulation of toxic products. RESULTS To further improve viral titers at high cell densities, perfusion culture mode was evaluated. Productivities of both perfusion and batch culture modes were compared at an infection cell density of 6 × 10(6) cells/mL. The metabolism, including glycolysis, glutaminolysis and amino acids utilization as well as physiological indicators such as viability and apoptosis were extensively documented for the two modes of culture before and after viral infection to identify potential metabolic limitations. A 3 L bioreactor with a perfusion rate of 0.5 vol/day allowed us to reach maximal titers of 3.3 × 10(11) IVP/mL and 4.0 logHA units/mL, corresponding to a total production of 1.0 × 10(15) IVP and 7.8 logHA units after 3 days post-infection. Overall, perfusion mode titers were higher by almost one order of magnitude over the batch culture mode of production. This improvement was associated with an activation of the cell metabolism as seen by a 1.5-fold and 4-fold higher consumption rates of glucose and glutamine respectively. A shift in the viral production kinetics was also observed leading to an accumulation of more viable cells with a higher specific production and causing an increase in the total volumetric production of infectious influenza particles. CONCLUSIONS These results confirm that the HEK293SF cell is an excellent substrate for high yield production of influenza virus. Furthermore, there is great potential in further improving the production yields through better control of the cell culture environment and viral production kinetics. Once accomplished, this cell line can be promoted as an industrial platform for cost-effective manufacturing of the influenza seasonal vaccine as well as for periods of peak demand during pandemics.
Collapse
Affiliation(s)
- Emma Petiot
- Biotechnology Research Institute, 6100 Royalmount Avenue, Montreal, H4P 2R2 Québec, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Ensuring the safety of vaccine cell substrates by massively parallel sequencing of the transcriptome. Vaccine 2011; 29:7117-21. [PMID: 21651935 DOI: 10.1016/j.vaccine.2011.05.071] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Massively parallel, deep, sequencing of the transcriptome coupled with algorithmic analysis to identify adventitious agents (MP-Seq™) is an important adjunct in ensuring the safety of cells used in vaccine production. Such cells may harbour novel viruses whose sequences are unknown or latent viruses that are only expressed following stress to the cells. MP-Seq is an unbiased and comprehensive method to identify such viruses and other adventitious agents without prior knowledge of the nature of those agents. Here we demonstrate its utility as part of an integrated approach to identify and characterise potential contaminants within commonly used virus and vaccine production cell lines. Through this analysis, in combination with more traditional approaches, we have excluded the presence of porcine circoviruses in the ATCC Vero cell bank (CCL-81), however, we found that a full length betaretrovirus related to SRV can be expressed in these cells, a factor that may be of importance in the production of certain vaccines. Similarly, insect cells are proving to be valuable for the production of virus like particles and sub-unit vaccines, but they can harbour a range of latent viruses. We show that following MP-Seq of the Trichoplusia ni (High Five cell line) transcriptome we were able to detect a contaminating, latent nodavirus and identify an expressed errantivirus genome. Collectively, these studies have reinforced the role of MP-Seq as an integral tool for the identification of contaminating agents in vaccine cell substrates.
Collapse
|
46
|
Gregersen JP, Schmitt HJ, Trusheim H, Bröker M. Safety of MDCK cell culture-based influenza vaccines. Future Microbiol 2011; 6:143-52. [DOI: 10.2217/fmb.10.161] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
After more than 60 years, the conventional production of influenza vaccines employing fertilized chicken eggs has reached its limits – both in terms of temporal flexibility and vaccine production volume. This problem is compounded by the fact that the pandemic-driven situation in 2009 has roughly doubled the overall vaccine demand. Modern cell culture technology has significant advantages over the conventional method of manufacturing influenza vaccines employing embryonated chicken eggs, and enables manufacturers to respond rapidly to the increasing worldwide seasonal and pandemic-driven need for influenza vaccines. Recent articles in the popular press claiming that cell culture-based influenza vaccines can cause tumors have fomented uncertainty among the general population and physicians, and also discredit officially accepted test results and product licensing. This article provides an overview of the safety profile of the cell culture technology, of the cells and of the final vaccine product.
Collapse
Affiliation(s)
- Jens-Peter Gregersen
- Novartis Vaccines & Diagnostics GmbH, Emil-von-Behring-Strasse 76, D-35041 Marburg, Germany
| | - Heinz-Josef Schmitt
- Novartis Vaccines & Diagnostics GmbH, Emil-von-Behring-Strasse 76, D-35041 Marburg, Germany
| | - Heidi Trusheim
- Novartis Vaccines & Diagnostics GmbH, Emil-von-Behring-Strasse 76, D-35041 Marburg, Germany
| | | |
Collapse
|