1
|
Cai M, Le Y, Gong Z, Dong T, Liu B, Su M, Li X, Peng F, Li Q, Nian X, Yu H, Wu Z, Zhang Z, Zhang J. Production, Passaging Stability, and Histological Analysis of Madin-Darby Canine Kidney Cells Cultured in a Low-Serum Medium. Vaccines (Basel) 2024; 12:991. [PMID: 39340023 PMCID: PMC11435615 DOI: 10.3390/vaccines12090991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Madin-Darby canine kidney (MDCK) cells are commonly used to produce cell-based influenza vaccines. However, the role of the low-serum medium on the proliferation of MDCK cells and the propagation of the influenza virus has not been well studied. In the present study, we used 5 of 15 culture methods with different concentrations of a mixed medium and neonatal bovine serum (NBS) to determine the best culture medium. We found that a VP:M199 ratio of 1:2 (3% NBS) was suitable for culturing MDCK cells. Furthermore, the stable growth of MDCK cells and the production of the influenza virus were evaluated over long-term passaging. We found no significant difference in terms of cell growth and virus production between high and low passages of MDCK cells under low-serum culture conditions, regardless of influenza virus infection. Lastly, we performed a comparison of the transcriptomics and proteomics of MDCK cells cultured in VP:M199 = 1:2 (3% NBS) with those cultured in VP:M199 = 1:2 (5% NBS) before and after influenza virus infection. The transcriptome analysis showed that differentially expressed genes were predominantly enriched in the metabolic pathway and MAPK signaling pathway, indicating an activated state. This suggests that decreasing the concentration of serum in the medium from 5% to 3% may increase the metabolic activity of cells. Proteomics analysis showed that only a small number of differentially expressed proteins could not be enriched for analysis, indicating minimal difference in the protein levels of MDCK cells when the serum concentration in the medium was decreased from 5% to 3%. Altogether, our findings suggest that the screening and application of a low-serum medium provide a background for the development and optimization of cell-based influenza vaccines.
Collapse
Affiliation(s)
- Ming Cai
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Yang Le
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zheng Gong
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Tianbao Dong
- Center for Drug Evaluation and Inspection of HMPA (Hubei Center for Vaccine Inspection), Wuhan 430207, China
| | - Bo Liu
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Minne Su
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuedan Li
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Feixia Peng
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Qingda Li
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Xuanxuan Nian
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Hao Yu
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zheng Wu
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Zhegang Zhang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| | - Jiayou Zhang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- National Key Laboratory for Novel Vaccines Research and Development of Emerging Infectious Diseases, Wuhan 430207, China
- Hubei Provincial Vaccine Technology Innovation Center, Wuhan 430207, China
| |
Collapse
|
2
|
Li H, Sun Z, Zheng T, Chen M, Lei X, Yu X, Ning Z. CD46 inhibits the replication of swine influenza viruses by promoting the production of type I IFNs in PK-15 cells. Vet Res Commun 2024; 48:1111-1119. [PMID: 38153594 DOI: 10.1007/s11259-023-10289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
Swine flu caused by swine influenza A virus (swIAV) is an acute respiratory viral disease that is spreading in swine herds worldwide. Although the effect of some host factors on replication of swIAV has been identified, the role of CD46 in this process is unclear. Here, we report that CD46 inhibits the replication of swIAV by promoting the production of type I interferons (IFNs) in porcine kidney (PK-15) cells. CD46 knockout (CD46-KO) and stably expressing (CD46-overexpression) PK-15 cells were prepared using lentivirus-mediated CRISPR/Cas9 gene editing and seamless cloning technology. The results of virus infection in CD46-overexpression PK-15 cells showed that the replication of H1N1 and H3N2 swIAVs were inhibited, and the production of type I IFNs (IFN-α, IFN-β), interferon regulatory factor (IRF) 3, and mitochondrial antiviral-signaling protein (MAVS) was enhanced. Virus infection in CD46-KO PK-15 cells showed the opposite results. Further results showed that CD46-KO PK-15 cells have a favorable ability to proliferate influenza viruses compared to Madin-Darby canine kidney (MDCK) and PK-15 cells. These findings indicate that CD46 acts as promising target regulating the replication of swIAV, and help to develop new agents against infection and replication of the virus.
Collapse
Affiliation(s)
- Huizi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenzhen Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Tingting Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ming Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoling Lei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xianglong Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhangyong Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
| |
Collapse
|
3
|
Yang Z, Yu S, Xu Y, Zhao Y, Li L, Sun J, Wang X, Guo Y, Zhang Y. The Screening and Mechanism of Influenza-Virus Sensitive MDCK Cell Lines for Influenza Vaccine Production. Diseases 2024; 12:20. [PMID: 38248371 PMCID: PMC10814076 DOI: 10.3390/diseases12010020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Influenza is a potentially fatal acute respiratory viral disease caused by the influenza virus. Influenza viruses vary in antigenicity and spread rapidly, resulting in seasonal epidemics. Vaccination is the most effective strategy for lowering the incidence and fatality rates of influenza-related disorders, and it is also an important method for reducing seasonal influenza infections. Mammalian Madin-Darby canine kidney (MDCK) cell lines are recommended for influenza virus growth, and such cell lines have been utilized in several commercial influenza vaccine productions. The limit dilution approach was used to screen ATCC-MDCK cell line subcellular strains that are especially sensitive to H1N1, H3N2, BV, and BY influenza viruses to increase virus production, and research on influenza virus culture media was performed to support influenza virus vaccine development. We also used RNA sequencing to identify differentially expressed genes and a GSEA analysis to determine the biological mechanisms underlying the various levels of susceptibility of cells to influenza viruses. MDCK cell subline 2B6 can be cultured to increase titer and the production of the H1N1, H3N2, BV, and BY influenza viruses. MDCK-2B6 has a significantly enriched and activated in ECM receptor interaction, JAK-STAT signaling, and cytokine receptor interaction signaling pathways, which may result in increased cellular susceptibility and cell proliferation activity to influenza viruses, promote viral adsorption and replication, and elevate viral production, ultimately. The study revealed that MDCK-2B6 can increase the influenza virus titer and yield in vaccine production by increasing cell sensitivity and enhancing proliferative activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100176, China; (Z.Y.); (Y.X.); (Y.Z.); (L.L.); (J.S.); (X.W.); (Y.G.)
| |
Collapse
|
4
|
Gupta D, Mohan S. Influenza vaccine: a review on current scenario and future prospects. J Genet Eng Biotechnol 2023; 21:154. [PMID: 38030859 PMCID: PMC10686931 DOI: 10.1186/s43141-023-00581-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023]
Abstract
Vaccination is a crucial tool in preventing influenza, but it requires annual updates in vaccine composition due to the ever-changing nature of the flu virus. While healthcare and economic burdens have reduced, the virus remains a challenge. Research conducted over the past decade has revealed pathways for improvement through both basic and clinical studies. Viral surveillance plays a vital role in the better selection of candidate viruses for vaccines and the early detection of drug-resistant strains.This page offers a description of future vaccine developments and an overview of current vaccine options. In the coming years, we anticipate significant changes in vaccine production, moving away from traditional egg-based methods towards innovative technologies such as DNA and RNA vaccines. These newer approaches offer significant advantages over traditional egg-based and cell culture-based influenza vaccine manufacturing.
Collapse
Affiliation(s)
- Dipanshi Gupta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh (AUUP), Sector-125, Noida, Uttar Pradesh, 201303, India
| | - Sumedha Mohan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh (AUUP), Sector-125, Noida, Uttar Pradesh, 201303, India.
| |
Collapse
|
5
|
Malenovská H. Ruxolitinib accelerates influenza A virus adaptation in the Madin-Darby canine kidney (MDCK) cell line. J Appl Microbiol 2023; 134:lxad232. [PMID: 37816667 DOI: 10.1093/jambio/lxad232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023]
Abstract
AIM To investigate the effect of ruxolitinib medium supplement, separately and in combination with trypsin, on influenza A virus (IAV) adaptation and propagation in the Madin-Darby canine kidney (MDCK) cell line. METHODS AND RESULTS Two consecutive passages of three egg-based IAV strains were performed in the MDCK cell line with medium (a) without additives; (b) with a combination of ruxolitinib and trypsin; (c) with ruxolitinib; and (d) trypsin. Adaptation without a medium additive failed in both passages. After a single passage, the probability of the IAV adaptation was highly significantly influenced by the type of additive (binomial generalized linear model, χ22 = 23.84, P < 0.00001). The highest probability of adaptation was achieved with the combination of ruxolitinib and trypsin, followed by ruxolitinib alone and trypsin. After the two consecutive passages, the influence of the type of medium additive on the probability of virus adaptation was no longer significant. In two of three IAV MDCK-adapted strains, the type of medium additive had no significant influence on virus yields. CONCLUSION Ruxolitinib accelerates the adaptation of IAV in the MDCK cell line both individually and together with trypsin.
Collapse
Affiliation(s)
- Hana Malenovská
- Collection of Animal Pathogenic Microorganisms, Veterinary Research Institute,Hudcova 296/70, 621 00 Brno-Medlánky, Czech Republic
| |
Collapse
|
6
|
Le Y, Zhang J, Gong Z, Zhang Z, Nian X, Li X, Yu D, Ma N, Zhou R, Zhang G, Liu B, Yang L, Fu B, Xu X, Yang X. TRAF3 deficiency in MDCK cells improved sensitivity to the influenza A virus. Heliyon 2023; 9:e19246. [PMID: 37681145 PMCID: PMC10481187 DOI: 10.1016/j.heliyon.2023.e19246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Tumor necrosis factor receptor-associated factor 3 (TRAF3), an adaptor protein, has significant and varying effects on immunity depending on cell types. The role of TRAF3 in Madin-Darby Canine Kidney Epithelial (MDCK) cell resistance to influenza A virus (IVA) remains elusive. In the present study, CRISPR-Cas9 gene editing technology was used to construct the TRAF3 knockout MDCK cells (MDCK-TRAF3-/-). Hemagglutination assay, plaque assay, transcriptome, and quantitative real-time PCR were performed after IVA infection. The results showed that after IVA infection, HA titers and virus titers were promoted, interferon I-related pathways were significantly blocked, and transcription of several antiviral-related genes was significantly decreased in MDCK-TRAF3-/- cells. Thus, our study suggests that TRAF3 gene knockout reduced MDCK cell's resistance to IVA, thereby resulting in a promising way for IVA isolation and vaccine manufacturing.
Collapse
Affiliation(s)
- Yang Le
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Zheng Gong
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Zhegang Zhang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Xuedan Li
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Daiguan Yu
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Ning Ma
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Rong Zhou
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Guomei Zhang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Bo Liu
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Lu Yang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Baiqi Fu
- Wuhan Institute of Biotechnology, Wuhan, 430075, China
| | - Xiuqin Xu
- Wuhan Institute of Biotechnology, Wuhan, 430075, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
- China National Biotech Group Company Limited, 100029, Bejing, China
| |
Collapse
|
7
|
Xu H, Zhu S, Govinden R, Chenia HY. Multiple Vaccines and Strategies for Pandemic Preparedness of Avian Influenza Virus. Viruses 2023; 15:1694. [PMID: 37632036 PMCID: PMC10459121 DOI: 10.3390/v15081694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Avian influenza viruses (AIV) are a continuous cause of concern due to their pandemic potential and devasting effects on poultry, birds, and human health. The low pathogenic avian influenza virus has the potential to evolve into a highly pathogenic avian influenza virus, resulting in its rapid spread and significant outbreaks in poultry. Over the years, a wide array of traditional and novel strategies has been implemented to prevent the transmission of AIV in poultry. Mass vaccination is still an economical and effective approach to establish immune protection against clinical virus infection. At present, some AIV vaccines have been licensed for large-scale production and use in the poultry industry; however, other new types of AIV vaccines are currently under research and development. In this review, we assess the recent progress surrounding the various types of AIV vaccines, which are based on the classical and next-generation platforms. Additionally, the delivery systems for nucleic acid vaccines are discussed, since these vaccines have attracted significant attention following their significant role in the fight against COVID-19. We also provide a general introduction to the dendritic targeting strategy, which can be used to enhance the immune efficiency of AIV vaccines. This review may be beneficial for the avian influenza research community, providing ideas for the design and development of new AIV vaccines.
Collapse
Affiliation(s)
- Hai Xu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Roshini Govinden
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Hafizah Y. Chenia
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
8
|
Deng X, Chen Z, Zhao Z, Chen J, Li M, Yang J, Yu H. Regional characteristics of influenza seasonality patterns in mainland China, 2005-2017: a statistical modeling study. Int J Infect Dis 2023; 128:91-97. [PMID: 36581188 DOI: 10.1016/j.ijid.2022.12.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/06/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES To quantify the seasonal and antigenic characteristics of influenza to help understand influenza activity and inform vaccine recommendations. METHODS We employed a generalized linear model with harmonic terms to quantify the seasonal pattern of influenza in China from 2005-2017, including amplitude (circulatory intensity), semiannual periodicity (given two peaks a year), annual peak time, and epidemic duration. The antigenic differences were distinguished as antigenic similarity between 2009 and 2020. We categorized regions above 33° N, between 27° N and 33° N, and below 27° N as the north, central, and south regions, respectively. RESULTS We estimated that the amplitude in the north region (median: 0.019, 95% CI: 0.018-0.021) was significantly higher than that in the central region (median: 0.011, 95% CI: 0.01-0.012, P <0.001) and south region (median: 0.008, 95% CI: 0.007-0.008, P <0.001) for influenza A virus subtype H3N2 (A/H3N2). The A/H3N2 in the central region had a semiannual periodicity (median: 0.548, 95% CI: 0.517-0.577), while no semiannual pattern was found in other regions or subtypes/lineages. The antigenic similarity was low (below 50% in the 2009-2010, 2014-2015, 2016-2018, and 2019-2020 seasons) for A/H3N2. CONCLUSION Our study depicted the seasonal pattern differences and antigenic differences of influenza in China, which provides information for vaccination strategies.
Collapse
Affiliation(s)
- Xiaowei Deng
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Zhiyuan Chen
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Zeyao Zhao
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Junbo Chen
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Mei Li
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Juan Yang
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Hongjie Yu
- Department of Infectious Diseases, Huashan Hospital, School of Public Health, Fudan University, Shanghai, China; Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China; National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Matsuda T, Tanijima T, Hirose A, Masumi-Koizumi K, Katsuda T, Yamaji H. Production of influenza virus-like particles using recombinant insect cells. Biochem Eng J 2020; 163:107757. [PMID: 32834743 PMCID: PMC7427601 DOI: 10.1016/j.bej.2020.107757] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/22/2020] [Accepted: 08/13/2020] [Indexed: 01/23/2023]
Abstract
Influenza A virus-like particles (VLPs) were produced using recombinant insect cells. VLPs were produced using insect cells as host cells without using a baculovirus. A secretory form of VLPs consists of hemagglutinin and matrix protein 1. The VLP productivity is comparable to that of the baculovirus–insect cell system.
Virus-like particles (VLPs) are hollow nanoparticles composed of recombinant viral surface proteins without a virus genome. In the present study, we investigated the production of influenza VLPs using recombinant insect cells. DNA fragments encoding influenza A virus hemagglutinin (HA) and matrix protein 1 (M1) were cloned with the Drosophila BiP signal sequence in plasmid vectors containing a blasticidin and a neomycin resistance gene, respectively. After Trichoplusia ni BTI-TN-5B1-4 (High Five) cells were co-transfected with a pair of constructed plasmid vectors, stably transformed cells were established via incubation with blasticidin and G418. Western blot analyses showed that recombinant High Five cells secreted HA and M1 proteins into the culture supernatant. Immunoprecipitation of the culture supernatant with an anti-HA antibody and transmission electron microscopy suggested that secreted HA and M1 proteins were in a particulate structure with a morphology similar to that of an influenza virus. Hemagglutination assay indicated that expressed HA molecules retained hemagglutination activity. In a shake-flask culture, recombinant cells achieved a high HA yield (≈ 10 μg/ml) comparable to the yields obtained using the baculovirus–insect cell system. Recombinant insect cells may serve as excellent platforms for the efficient production of influenza VLPs for use as safe and effective vaccines and diagnostic antigens.
Collapse
Affiliation(s)
- Takuya Matsuda
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Toshikazu Tanijima
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Akito Hirose
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Kyoko Masumi-Koizumi
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Tomohisa Katsuda
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Hideki Yamaji
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| |
Collapse
|
10
|
Yamayoshi S, Kawaoka Y. Current and future influenza vaccines. Nat Med 2019; 25:212-220. [PMID: 30692696 DOI: 10.1038/s41591-018-0340-z] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/19/2018] [Indexed: 11/09/2022]
Abstract
Although antiviral drugs and vaccines have reduced the economic and healthcare burdens of influenza, influenza epidemics continue to take a toll. Over the past decade, research on influenza viruses has revealed a potential path to improvement. The clues have come from accumulated discoveries from basic and clinical studies. Now, virus surveillance allows researchers to monitor influenza virus epidemic trends and to accumulate virus sequences in public databases, which leads to better selection of candidate viruses for vaccines and early detection of drug-resistant viruses. Here we provide an overview of current vaccine options and describe efforts directed toward the development of next-generation vaccines. Finally, we propose a plan for the development of an optimal influenza vaccine.
Collapse
Affiliation(s)
- Seiya Yamayoshi
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan. .,Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan. .,Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin Madison, Madison, WI, USA.
| |
Collapse
|
11
|
Melinek BJ, Dessoy S, Wright B, Bracewell DG, Mukhopadhyay TK. Ultra scale-down approaches to study the centrifugal harvest for viral vaccine production. Biotechnol Bioeng 2018; 115:1226-1238. [PMID: 29315484 DOI: 10.1002/bit.26546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 01/03/2018] [Indexed: 02/06/2023]
Abstract
Large scale continuous cell-line cultures promise greater reproducibility and efficacy for the production of influenza vaccines, and adenovirus for gene therapy. This paper seeks to use an existing validated ultra scale-down tool, which is designed to mimic the commercial scale process environment using only milliliters of material, to provide some initial insight into the performance of the harvest step for these processes. The performance of industrial scale centrifugation and subsequent downstream process units is significantly affected by shear. The properties of these cells, in particular their shear sensitivity, may be changed considerably by production of a viral product, but literature on this is limited to date. In addition, the scale-down tool used here has not previously been applied to the clarification of virus production processes. The results indicate that virus infected cells do not actually show any increase in sensitivity to shear, and may indeed become less shear sensitive, in a similar manner to that previously observed in old or dead cell cultures. Clarification may be most significantly dependent on the virus release mechanism, with the budding influenza virus producing a much greater decrease in clarification than the lytic, non-enveloped adenovirus. A good match was also demonstrated to the industrial scale performance in terms of clarification, protein release, and impurity profile.
Collapse
Affiliation(s)
- Beatrice J Melinek
- Department of Biochemical Engineering, Bernard Katz building, University College London, London, UK
| | | | - Bernice Wright
- Department of Biochemical Engineering, Bernard Katz building, University College London, London, UK
| | - Dan G Bracewell
- Department of Biochemical Engineering, Bernard Katz building, University College London, London, UK
| | - Tarit K Mukhopadhyay
- Department of Biochemical Engineering, Bernard Katz building, University College London, London, UK
| |
Collapse
|
12
|
Safety and immunogenicity of an inactivated cell culture-derived H7N9 influenza vaccine in healthy adults: A phase I/II, prospective, randomized, open-label trial. Vaccine 2017; 35:4099-4104. [PMID: 28668573 DOI: 10.1016/j.vaccine.2017.06.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/26/2017] [Accepted: 06/19/2017] [Indexed: 01/27/2023]
Abstract
BACKGROUND We conducted a phase I/II clinical trial to evaluate the safety and immunogenicity of a Madin-Darby canine kidney (MDCK) cell-grown inactivated H7N9 influenza vaccine for pandemic preparedness purposes. METHODS Between April 7, 2015 and May 27, 2016, healthy adults aged 20-60years were enrolled sequentially in phase I (n=40) and phase II (n=160) from three hospitals in Taiwan and randomized to receive 2 doses of whole-virus H7N9 vaccine (15 or 30μg hemagglutinin antigen (HA) with or without an aluminum hydroxide adjuvant) at 21-day intervals. Safety up to 180days and changes in hemagglutinin inhibition (HI) titers at 21days after each vaccination were determined. RESULTS Of the 200 randomized subjects, 193 (96.5%) received 2 doses of the study vaccine and were included in the intention-to-treat analysis for safety, and 190 (95%) were included in the per-protocol analysis for immunogenicity. Most adverse events were mild and transient; no death or vaccine-related serious adverse events were reported. Overall, higher immune responses were observed in the groups administered with 30μgHA formulation than in the other two groups administered with 15μgHA formulation. The highest immune response was observed in subjects who received 2 doses of the adjuvanted vaccine containing 30μgHA with HI titer, seroprotection rate, seroconversion rate, and seroconversion factor of 36.2, 64.6%, 64.6% and 5.7, respectively. CONCLUSIONS Our study demonstrated that the H7N9 influenza vaccine containing 30µgHA with aluminum hydroxide adjuvant was immunogenic and safe in adults aged 20-60years. CLINICALTRIALS.GOV identifier: NCT02436928.
Collapse
|
13
|
Establishment of MDCK/FX Cell for Efficient Replication of Influenza Viruses. Jundishapur J Microbiol 2017. [DOI: 10.5812/jjm.44891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
14
|
Kang K, Han S, Hong T, Jeon S, Paek J, Kang JH, Yim DS. Immunogenicity and Safety of Trivalent Split Influenza Vaccine in Healthy Korean Adults with Low Pre-Existing Antibody Levels: An Open Phase I Trial. Yonsei Med J 2016; 57:1354-60. [PMID: 27593862 PMCID: PMC5011266 DOI: 10.3349/ymj.2016.57.6.1354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 12/02/2022] Open
Abstract
PURPOSE A phase I clinical trial was conducted to evaluate the immunogenicity and safety of newly developed egg-cultivated trivalent inactivated split influenza vaccine (TIV) in Korea. MATERIALS AND METHODS The TIV was administered to 43 healthy male adults. Subjects with high pre-existing titers were excluded in a screening step. Immune response was measured by a hemagglutination inhibition (HI) assay. RESULTS The seroprotection rates against A/California/7/2009 (H1N1), A/Perth/16/2009 (H3N2) and B/Brisbane/60/2009 were 74.42% [95% confidence interval (CI): 61.38-87.46], 72.09% (95% CI: 58.69-85.50), and 86.05% (95% CI: 75.69-96.40), respectively. Calculated seroconversion rates were 74.42% (95% CI: 61.38-87.46), 74.42% (95% CI: 61.38-87.46), and 79.07% (95% CI: 66.91-91.23), respectively. There were 25 episodes of solicited local adverse events in 21 subjects (47.73%), 21 episodes of solicited general adverse events in 16 subjects (36.36%) and 5 episodes of unsolicited adverse events in 5 subjects (11.36%). All adverse events were grade 1 or 2 and disappeared within three days. CONCLUSION The immunogenicity and safety of TIV established in this phase I trial are sufficient to plan a larger scale clinical trial.
Collapse
Affiliation(s)
- Kyuri Kang
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seunghoon Han
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Taegon Hong
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sangil Jeon
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeongki Paek
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Han Kang
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dong Seok Yim
- Department of Clinical Pharmacology and Therapeutics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
15
|
Chen H, Deng Q, Ng SH, Lee RTC, Maurer-Stroh S, Zhai W. Dynamic Convergent Evolution Drives the Passage Adaptation across 48 Years' History of H3N2 Influenza Evolution. Mol Biol Evol 2016; 33:3133-3143. [PMID: 27604224 DOI: 10.1093/molbev/msw190] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Influenza viruses are often propagated in a diverse set of culturing media and additional substitutions known as passage adaptation can cause extra evolution in the target strain, leading to ineffective vaccines. Using 25,482 H3N2 HA1 sequences curated from Global Initiative on Sharing All Influenza Data and National Center for Biotechnology Information databases, we found that passage adaptation is a very dynamic process that changes over time and evolves in a seesaw like pattern. After crossing the species boundary from bird to human in 1968, the influenza H3N2 virus evolves to be better adapted to the human environment and passaging them in embryonated eggs (i.e., an avian environment) leads to increasingly stronger positive selection. On the contrary, passage adaptation to the mammalian cell lines changes from positive selection to negative selection. Using two statistical tests, we identified 19 codon positions around the receptor binding domain strongly contributing to passage adaptation in the embryonated egg. These sites show strong convergent evolution and overlap extensively with positively selected sites identified in humans, suggesting that passage adaptation can confound many of the earlier studies on influenza evolution. Interestingly, passage adaptation in recent years seems to target a few codon positions in antigenic surface epitopes, which makes it difficult to produce antigenically unaltered vaccines using embryonic eggs. Our study outlines another interesting scenario whereby both convergent and adaptive evolution are working in synchrony driving viral adaptation. Future studies from sequence analysis to vaccine production need to take careful consideration of passage adaptation.
Collapse
Affiliation(s)
- Hui Chen
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore
| | - Qiang Deng
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore.,Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Sebastian Maurer-Stroh
- Bioinformatics Institute, A*STAR, Singapore.,School of Biological Sciences (SBS), Nanyang Technological University (NTU), Singapore.,National Public Health Laboratory (NPHL), Ministry of Health (MOH), Singapore.,Department of Biological Sciences, National University of Singapore (NUS), Singapore
| | - Weiwei Zhai
- Human Genetics, Genome Institute of Singapore, A*STAR, Singapore
| |
Collapse
|
16
|
Zahoor MA, Khurshid M, Qureshi R, Naz A, Shahid M. Cell culture-based viral vaccines: current status and future prospects. Future Virol 2016. [DOI: 10.2217/fvl-2016-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cell culture-based viral vaccines are used globally to immunize humans against infections. The cell culture is continuous process of developing substrates for the safe production of viral vaccines. However, increased global demand and strict safety rules for novel vaccines to control and eradicate viral diseases have forced researchers and manufacturers toward cell culture-based vaccines. The choice of cell substrate is a critical step that cannot be generalized for every vaccine formulation, therefore, manufacturers intend to optimize the required processes for particular applications. The recently established cell lines, innovative bioreactor concepts and cultivation schemes are necessary to increase the potential of vaccine production. In this review, we have focused on current cell culture-based viral vaccines and their future prospects.
Collapse
Affiliation(s)
| | - Mohsin Khurshid
- Department of Microbiology, Government College University, Faisalabad, Pakistan
- College of Allied Health Professionals, Directorate of Medical Sciences, Government College University, Faisalabad, Pakistan
| | - Rabia Qureshi
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Aneeqa Naz
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Muhammad Shahid
- Department of Bioinformatics & Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
17
|
Wörmann X, Lesch M, Welke RW, Okonechnikov K, Abdurishid M, Sieben C, Geissner A, Brinkmann V, Kastner M, Karner A, Zhu R, Hinterdorfer P, Anish C, Seeberger PH, Herrmann A, Meyer TF, Karlas A. Genetic characterization of an adapted pandemic 2009 H1N1 influenza virus that reveals improved replication rates in human lung epithelial cells. Virology 2016; 492:118-29. [PMID: 26914510 DOI: 10.1016/j.virol.2016.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/03/2015] [Accepted: 02/03/2016] [Indexed: 12/22/2022]
Abstract
The 2009 influenza pandemic originated from a swine-origin H1N1 virus, which, although less pathogenic than anticipated, may acquire additional virulence-associated mutations in the future. To estimate the potential risk, we sequentially passaged the isolate A/Hamburg/04/2009 in A549 human lung epithelial cells. After passage 6, we observed a 100-fold increased replication rate. High-throughput sequencing of viral gene segments identified five dominant mutations, whose contribution to the enhanced growth was analyzed by reverse genetics. The increased replication rate was pinpointed to two mutations within the hemagglutinin (HA) gene segment (HA1 D130E, HA2 I91L), near the receptor binding site and the stem domain. The adapted virus also replicated more efficiently in mice in vivo. Enhanced replication rate correlated with increased fusion pH of the HA protein and a decrease in receptor affinity. Our data might be relevant for surveillance of pre-pandemic strains and development of high titer cell culture strains for vaccine production.
Collapse
Affiliation(s)
- Xenia Wörmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Markus Lesch
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Steinbeis Innovation gGmbH, Center for Systems Biomedicine, Falkensee, Germany
| | - Robert-William Welke
- Department of Biology, Molecular Biophysics, IRI Life Sciences, Humboldt-Universität zu Berlin, Germany
| | - Konstantin Okonechnikov
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Mirshat Abdurishid
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Christian Sieben
- Department of Biology, Molecular Biophysics, IRI Life Sciences, Humboldt-Universität zu Berlin, Germany
| | - Andreas Geissner
- Department for Biomolecular Systems, Max Planck Institute for Colloids and Interfaces, Potsdam, Germany; Institute of Chemistry and Biochemistry, Free University, Berlin, Germany
| | - Volker Brinkmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Markus Kastner
- Institute for Biophysics, Johannes Kepler University, Linz, Austria
| | - Andreas Karner
- Center for Advanced Bioanalysis GmbH (CBL), Linz, Austria
| | - Rong Zhu
- Institute for Biophysics, Johannes Kepler University, Linz, Austria
| | | | - Chakkumkal Anish
- Department for Biomolecular Systems, Max Planck Institute for Colloids and Interfaces, Potsdam, Germany
| | - Peter H Seeberger
- Department for Biomolecular Systems, Max Planck Institute for Colloids and Interfaces, Potsdam, Germany; Institute of Chemistry and Biochemistry, Free University, Berlin, Germany
| | - Andreas Herrmann
- Department of Biology, Molecular Biophysics, IRI Life Sciences, Humboldt-Universität zu Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Steinbeis Innovation gGmbH, Center for Systems Biomedicine, Falkensee, Germany.
| | - Alexander Karlas
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Steinbeis Innovation gGmbH, Center for Systems Biomedicine, Falkensee, Germany.
| |
Collapse
|
18
|
Steil D, Schepers CL, Pohlentz G, Legros N, Runde J, Humpf HU, Karch H, Müthing J. Shiga toxin glycosphingolipid receptors of Vero-B4 kidney epithelial cells and their membrane microdomain lipid environment. J Lipid Res 2015; 56:2322-36. [PMID: 26464281 DOI: 10.1194/jlr.m063040] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Indexed: 12/12/2022] Open
Abstract
Shiga toxins (Stxs) are produced by enterohemorrhagic Escherichia coli (EHEC), which cause human infections with an often fatal outcome. Vero cell lines, derived from African green monkey kidney, represent the gold standard for determining the cytotoxic effects of Stxs. Despite their global use, knowledge about the exact structures of the Stx receptor glycosphingolipids (GSLs) and their assembly in lipid rafts is poor. Here we present a comprehensive structural analysis of Stx receptor GSLs and their distribution to detergent-resistant membranes (DRMs), which were prepared from Vero-B4 cells and used as lipid raft equivalents. We identified globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer) as the GSL receptors for Stx1a, Stx2a, and Stx2e subtypes using TLC overlay detection combined with MS. The uncommon Stx receptor, globopentaosylceramide (Gb5Cer, Galβ3GalNAcβ3Galα4Galβ4Glcβ1Cer), which was specifically recognized (in addition to Gb3Cer and Gb4Cer) by Stx2e, was fully structurally characterized. Lipoforms of Stx receptor GSLs were found to mainly harbor ceramide moieties composed of sphingosine (d18:1) and C24:0/C24:1 or C16:0 fatty acid. Moreover, co-occurrence with lipid raft markers, SM and cholesterol, in DRMs suggested GSL association with membrane microdomains. This study provides the basis for further exploring the functional impact of lipid raft-associated Stx receptors for toxin-mediated injury of Vero-B4 cells.
Collapse
Affiliation(s)
- Daniel Steil
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany
| | | | | | - Nadine Legros
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany
| | - Jana Runde
- Food Chemistry, University of Münster, D-48149 Münster, Germany
| | | | - Helge Karch
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institutes for Hygiene University of Münster, D-48149 Münster, Germany Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany
| |
Collapse
|
19
|
Zhang H, Han Q, Ping X, Li L, Chang C, Chen Z, Shu Y, Xu K, Sun B. A single NS2 mutation of K86R promotes PR8 vaccine donor virus growth in Vero cells. Virology 2015; 482:32-40. [PMID: 25817403 DOI: 10.1016/j.virol.2015.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 12/30/2014] [Accepted: 03/02/2015] [Indexed: 02/05/2023]
Abstract
Vaccination is the most effective way to prevent and control infection by influenza viruses, and a cell-culture-based vaccine production system is preferred as the future choice for the large-scale production of influenza vaccines. As one of the WHO-recommended cell lines for producing influenza vaccines, Vero cells do not efficiently support the growth of the current influenza A virus vaccine donor strain, the A/Puerto Rico/8/1934 (PR8) virus. In this study, a single mutation of K86R in the NS2 protein can sufficiently render the high-yielding property to the PR8 virus in Vero cells. Further analysis showed that the later steps in the virus replication cycle were accelerated by NS2(K86R) mutation, which may relate to an enhanced interaction between NS2(K86R) and the components of host factor F1Fo-ATPase, FoB and F1β. Because the NS2(K86R) mutation does not increase PR8 virulence in either mice or embryonated eggs, the PR8-NS2(K86R) virus could serve as a promising vaccine donor strain in Vero cells.
Collapse
Affiliation(s)
- Hong Zhang
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Qinglin Han
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Xianqiang Ping
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China; Shanghai Normal University, No. 100 Guilin Road, Shanghai 200234, China
| | - Li Li
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Chong Chang
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Ze Chen
- Shanghai Institute of Biological Products, Shanghai 200052, China
| | - Yuelong Shu
- Chinese Center for Disease Control and Prevention, Yingxin Street 100, Xuanwu District, Beijing 100052, China
| | - Ke Xu
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China.
| | - Bing Sun
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China; State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China.
| |
Collapse
|
20
|
Schuind A, Segall N, Drame M, Innis BL. Immunogenicity and Safety of an EB66 Cell-Culture-Derived Influenza A/Indonesia/5/2005(H5N1) AS03-Adjuvanted Vaccine: A Phase 1 Randomized Trial. J Infect Dis 2015; 212:531-41. [PMID: 25722291 PMCID: PMC4512608 DOI: 10.1093/infdis/jiv091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 02/09/2015] [Indexed: 01/08/2023] Open
Abstract
Background. Cell-culture-derived (CC) influenza vaccine production methods could provide benefits over classical embryonated-egg technology, including a higher production capacity and the faster creation of a supply that meets demand. Methods. A CC-inactivated split-virus influenza A/Indonesia/5/2005(H5N1) vaccine derived from the EB66 cell line (hereafter, “CC-H5N1”) was investigated in a phase 1 randomized, blinded study. Healthy adults (n = 521) received 2 vaccine doses (days 0 and 21) of either investigational CC-H5N1 vaccine (1.9 µg or 3.75 µg of hemagglutinin antigen [HA] with the AS03 adjuvant system or 15 µg of plain HA), embryonated-egg-derived vaccines (3.75 µg of HA with AS03 or 15 µg of plain HA), or placebo. Assessment of the adjuvant effect and immunogenicity was performed using Center for Biologics Evaluation and Research acceptability criteria 21 days after dose 2. Safety was assessed until month 12. Results. AS03-adjuvanted CC-H5N1 elicited a homologous hemagglutination inhibition antibody response that satisfied immunogenicity criteria 21 days after dose 2 and persisted at month 12. Adjuvant effect and immune response against a drift-variant strain were demonstrated. No vaccine-related serious adverse events were reported. The immunogenicity and safety of the CC-H5N1 formulation containing 3.75 µg of HA and AS03 appeared to be similar to those for the licensed egg-derived AS03-adjuvanted control vaccine. Conclusions. The feasibility of the EB66 cell line to produce an immunogenic influenza vaccine with acceptable safety profile was demonstrated. Antigen sparing was achieved through combination with AS03 adjuvant. This CC-H5N1 might contribute to the rapid access of vaccine in the event of an influenza A(H5N1) pandemic. Clinical Trials Registration NCT01236040.
Collapse
|
21
|
Shahiduzzaman M, Ezatti P, Xin G, Coombs KM. Proteasomal Serine Hydrolases Are Up-Regulated by and Required for Influenza Virus Infection. J Proteome Res 2014; 13:2223-38. [DOI: 10.1021/pr5001779] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Md Shahiduzzaman
- Department
of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | | | - Gang Xin
- Department
of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
- Department
of Microbiology and Immunology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Kevin M. Coombs
- Department
of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| |
Collapse
|
22
|
Yamaji H. Suitability and perspectives on using recombinant insect cells for the production of virus-like particles. Appl Microbiol Biotechnol 2014; 98:1963-70. [PMID: 24407451 DOI: 10.1007/s00253-013-5474-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/12/2013] [Accepted: 12/14/2013] [Indexed: 10/25/2022]
Abstract
Virus-like particles (VLPs) can be produced in recombinant protein production systems by expressing viral surface proteins that spontaneously assemble into particulate structures similar to authentic viral or subviral particles. VLPs serve as excellent platforms for the development of safe and effective vaccines and diagnostic antigens. Among various recombinant protein production systems, the baculovirus-insect cell system has been used extensively for the production of a wide variety of VLPs. This system is already employed for the manufacture of a licensed human papillomavirus-like particle vaccine. However, the baculovirus-insect cell system has several inherent limitations including contamination of VLPs with progeny baculovirus particles. Stably transformed insect cells have emerged as attractive alternatives to the baculovirus-insect cell system. Different types of VLPs, with or without an envelope and composed of either single or multiple structural proteins, have been produced in stably transformed insect cells. VLPs produced by stably transformed insect cells have successfully elicited immune responses in vivo. In some cases, the yield of VLPs attained with recombinant insect cells was comparable to, or higher than, that obtained by baculovirus-infected insect cells. Recombinant insect cells offer a promising approach to the development and production of VLPs.
Collapse
Affiliation(s)
- Hideki Yamaji
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657-8501, Japan,
| |
Collapse
|
23
|
Perdue ML, Arnold F, Li S, Donabedian A, Cioce V, Warf T, Huebner R. The future of cell culture-based influenza vaccine production. Expert Rev Vaccines 2014; 10:1183-94. [DOI: 10.1586/erv.11.82] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
24
|
Genzel Y, Rödig J, Rapp E, Reichl U. Vaccine production: upstream processing with adherent or suspension cell lines. Methods Mol Biol 2014; 1104:371-393. [PMID: 24297427 DOI: 10.1007/978-1-62703-733-4_23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The production of viral vaccines in cell culture can be accomplished with primary, diploid, or continuous (transformed) cell lines. Each cell line, each virus type, and each vaccine preparation require the specific design of upstream and downstream processing. Media have to be selected as well as production vessels, cultivation conditions, and modes of operation. Many viruses only replicate to high titers in adherently growing cells, but similar to processes established for recombinant protein production, an increasing number of suspension cell lines is being evaluated for future use. Here, we describe key issues to be considered for the establishment of large-scale virus production in bioreactors. As an example upstream processing of cell culture-derived influenza virus production is described in more detail for adherently growing and for suspension cells. In particular, use of serum-containing, serum-free, and chemically defined media as well as choice of cultivation vessel are considered.
Collapse
Affiliation(s)
- Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | | | | | | |
Collapse
|
25
|
Comparison of influenza virus yields and apoptosis-induction in an adherent and a suspension MDCK cell line. Vaccine 2013; 31:5693-9. [DOI: 10.1016/j.vaccine.2013.09.051] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/22/2013] [Accepted: 09/24/2013] [Indexed: 01/09/2023]
|
26
|
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: 39] [Impact Index Per Article: 3.5] [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
|
27
|
Yamaji H, Konishi E. Production of Japanese encephalitis virus-like particles in insect cells. Bioengineered 2013; 4:438-42. [PMID: 23639981 DOI: 10.4161/bioe.24514] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Virus-like particles (VLPs) are composed of one or several recombinant viral surface proteins that spontaneously assemble into particulate structures without the incorporation of virus DNA or RNA. The baculovirus-insect cell system has been used extensively for the production of recombinant virus proteins including VLPs. While the baculovirus-insect cell system directs the transient expression of recombinant proteins in a batch culture, stably transformed insect cells allow constitutive production. In our recent study, a secretory form of Japanese encephalitis (JE) VLPs was successfully produced by Trichoplusia ni BTI-TN-5B1-4 (High Five) cells engineered to coexpress the JE virus (JEV) premembrane (prM) and envelope (E) proteins. A higher yield of E protein was attained with recombinant High Five cells than with the baculovirus-insect cell system. This study demonstrated that recombinant insect cells offer a promising approach to the high-level production of VLPs for use as vaccines and diagnostic antigens.
Collapse
Affiliation(s)
- Hideki Yamaji
- Department of Chemical Science and Engineering; Graduate School of Engineering; Kobe University; Kobe, Japan; BIKEN Endowed Department of Dengue Vaccine Development; Faculty of Tropical Medicine; Mahidol University; Bangkok, Thailand
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development; Faculty of Tropical Medicine; Mahidol University; Bangkok, Thailand
| |
Collapse
|
28
|
Optimization of Insect Cell Based Protein Production Processes - Online Monitoring, Expression Systems, Scale Up. YELLOW BIOTECHNOLOGY II 2013; 136:65-100. [DOI: 10.1007/10_2013_205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
29
|
Lohr V, Genzel Y, Jordan I, Katinger D, Mahr S, Sandig V, Reichl U. Live attenuated influenza viruses produced in a suspension process with avian AGE1.CR.pIX cells. BMC Biotechnol 2012; 12:79. [PMID: 23110398 PMCID: PMC3505166 DOI: 10.1186/1472-6750-12-79] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 10/15/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Current influenza vaccines are trivalent or quadrivalent inactivated split or subunit vaccines administered intramuscularly, or live attenuated influenza vaccines (LAIV) adapted to replicate at temperatures below body temperature and administered intranasally. Both vaccines are considered safe and efficient, but due to differences in specific properties may complement each other to ensure reliable vaccine coverage. By now, licensed LAIV are produced in embryonated chicken eggs. In the near future influenza vaccines for human use will also be available from adherent MDCK or Vero cell cultures, but a scalable suspension process may facilitate production and supply with vaccines. RESULTS We evaluated the production of cold-adapted human influenza virus strains in the duck suspension cell line AGE1.CR.pIX using a chemically-defined medium. One cold-adapted A (H1N1) and one cold-adapted B virus strain was tested, as well as the reference strain A/PR/8/34 (H1N1). It is shown that a medium exchange is not required for infection and that maximum virus titers are obtained for 1 × 10⁻⁶ trypsin units per cell. 1 L bioreactor cultivations showed that 4 × 10⁶ cells/mL can be infected without a cell density effect achieving titers of 1 × 10⁸ virions/mL after 24 h. CONCLUSIONS Overall, this study demonstrates that AGE1.CR.pIX cells support replication of LAIV strains in a chemically-defined medium using a simple process without medium exchanges. Moreover, the process is fast with peak titers obtained 24 h post infection and easily scalable to industrial volumes as neither microcarriers nor medium replacements are required.
Collapse
Affiliation(s)
- Verena Lohr
- Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Ingo Jordan
- ProBioGen AG, Goethestr. 54, 13086, Berlin, Germany
| | - Dietmar Katinger
- Polymun Scientific GmbH, Donaustr. 99, 3400, Klosterneuburg, Austria
| | - Stefan Mahr
- Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
- University for Applied Sciences, Robert-Gerwig-Platz 1, 78120, Furtwangen, Germany
| | | | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
- Chair of Bioprocess Engineering, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| |
Collapse
|
30
|
Isken B, Genzel Y, Reichl U. Productivity, apoptosis, and infection dynamics of influenza A/PR/8 strains and A/PR/8-based reassortants. Vaccine 2012; 30:5253-61. [PMID: 22698452 DOI: 10.1016/j.vaccine.2012.05.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 05/16/2012] [Accepted: 05/25/2012] [Indexed: 01/18/2023]
Abstract
In cell culture-based influenza vaccine production significant efforts are directed towards virus seed optimization for maximum yields. Typically, high growth reassortants (HGR) containing backbones of six gene segments of e.g. influenza A/PR/8, are generated from wild type strains. Often, however, HA and TCID₅₀ titres obtained do not meet expectations and further optimization measures are required. Flow cytometry is an invaluable tool to improve our understanding of mechanism related to progress of infection, virus-induced apoptosis, and cell-specific productivity. In this study, we performed infections with two influenza A/PR/8 variants (from NIBSC and RKI) and two A/PR/8-based HGRs (Wisconsin-like and Uruguay-like) to investigate virus replication, apoptosis and virus titres at different multiplicities of infection (MOI 0.0001, 0.1, 3). Flow cytometric analyses showed similar dynamics in the time course of infected and apoptotic cell populations for all four tested strains at MOI 0.0001. Interestingly, higher MOI resulted in an earlier increase of the populations of infected and apoptotic cells and showed strain-specific differences. Infections with A/PR/8 NIBSC resulted in an earlier increase in both cell populations compared to A/PR/8 RKI. The Uruguay-like reassortant showed the earliest increase in the concentration of infected cells and a late induction of apoptosis at all tested MOIs. In contrast, the Wisconsin-like reassortant showed strong apoptosis induction at high MOIs resulting in reduced titres compared to lower MOI. Maximum HA titres were unaffected by changes in the MOI for the two A/PR/8 and the Uruguay-like reassortant. Maximum TCID₅₀ titres, however, decreased with increasing MOI for all strains. Overall, infections at very low MOI (0.0001) resulted not only in similar dynamics concerning progress of infection and induction of apoptosis but also in maximum virus yields. Highest HA titres were obtained for virus seed strains combining a fast progress in infection with a late onset of apoptosis. Therefore, both factors should be considered for the establishment of robust influenza vaccine production processes.
Collapse
Affiliation(s)
- B Isken
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstrasse 1, 39106 Magdeburg, Germany.
| | | | | |
Collapse
|