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
|
Zheng Z, Chen B, Liu X, Guo R, Chi H, Chen X, Pan Y, Gong H. Novel Eel Skin Fibroblast Cell Line: Bridging Adherent and Suspension Growth for Aquatic Applications Including Virus Susceptibility. BIOLOGY 2024; 13:1068. [PMID: 39765736 PMCID: PMC11673813 DOI: 10.3390/biology13121068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Suspension growth can greatly increase the cell density and yield of cell metabolites. To meet the requirements of aquatic industries, a culture model derived from Anguilla anguilla skin was developed using the explant outgrowth and enzyme-digesting passaging methods. These cells were kept in vitro continuously for over 12 months and subcultured 68 times. This heteroploid cell line, designated as ES, can naturally adapt to adherent and suspension growth reversibly under certain temperatures, serum percentages, and inoculum densities, without the need for any microcarriers or special medium additives. The ES cells can continue being highly productive under a temperature range of 15-37 °C and a serum percentage ranging from 3 to 15%. An inoculum density higher than 5 × 105 cells·mL-1 is necessary for the ES cells to turn into suspension efficiently. The green fluorescent protein (GFP) reporter gene was successfully expressed in the ES cells. The ES cells demonstrated susceptibility to Anguillid herpesvirus (AngHV) and red-spotted grouper nervous necrosis virus (RGNNV). ES is the first natural suspension growth model of aquatic origin; it does not require the processes of suspension domestication and carrier dissolution, making it a promising and cost-effective model for vaccine production, bio-pharmaceutical manufacturing, and cellular agriculture.
Collapse
Affiliation(s)
- Zaiyu Zheng
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China; (Z.Z.); (B.C.); (X.L.); (H.C.); (X.C.); (Y.P.)
| | - Bin Chen
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China; (Z.Z.); (B.C.); (X.L.); (H.C.); (X.C.); (Y.P.)
| | - Xiaodong Liu
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China; (Z.Z.); (B.C.); (X.L.); (H.C.); (X.C.); (Y.P.)
| | - Rui Guo
- Fuzhou Ocean and Fisheries Technology Center, Fuzhou 350007, China
| | - Hongshu Chi
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China; (Z.Z.); (B.C.); (X.L.); (H.C.); (X.C.); (Y.P.)
| | - Xiuxia Chen
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China; (Z.Z.); (B.C.); (X.L.); (H.C.); (X.C.); (Y.P.)
| | - Ying Pan
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China; (Z.Z.); (B.C.); (X.L.); (H.C.); (X.C.); (Y.P.)
| | - Hui Gong
- Biotechnology Institute, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China; (Z.Z.); (B.C.); (X.L.); (H.C.); (X.C.); (Y.P.)
| |
Collapse
|
3
|
Demirden SF, Kimiz-Gebologlu I, Oncel SS. Animal Cell Lines as Expression Platforms in Viral Vaccine Production: A Post Covid-19 Perspective. ACS OMEGA 2024; 9:16904-16926. [PMID: 38645343 PMCID: PMC11025085 DOI: 10.1021/acsomega.3c10484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/23/2024]
Abstract
Vaccines are considered the most effective tools for preventing diseases. In this sense, with the Covid-19 pandemic, the effects of which continue all over the world, humanity has once again remembered the importance of the vaccine. Also, with the various epidemic outbreaks that occurred previously, the development processes of effective vaccines against these viral pathogens have accelerated. By these efforts, many different new vaccine platforms have been approved for commercial use and have been introduced to the commercial landscape. In addition, innovations have been made in the production processes carried out with conventionally produced vaccine types to create a rapid response to prevent potential epidemics or pandemics. In this situation, various cell lines are being positioned at the center of the production processes of these new generation viral vaccines as expression platforms. Therefore, since the main goal is to produce a fast, safe, and effective vaccine to prevent the disease, in addition to existing expression systems, different cell lines that have not been used in vaccine production until now have been included in commercial production for the first time. In this review, first current viral vaccine types in clinical use today are described. Then, the reason for using cell lines, which are the expression platforms used in the production of these viral vaccines, and the general production processes of cell culture-based viral vaccines are mentioned. Also, selection parameters for animal cell lines as expression platforms in vaccine production are explained by considering bioprocess efficiency and current regulations. Finally, all different cell lines used in cell culture-based viral vaccine production and their properties are summarized, with an emphasis on the current and future status of cell cultures in industrial viral vaccine production.
Collapse
Affiliation(s)
| | | | - Suphi S. Oncel
- Ege University, Bioengineering Department, Izmir, 35100, Turkiye
| |
Collapse
|
4
|
Saengchoowong S, Nimsamer P, Khongnomnan K, Poomipak W, Praianantathavorn K, Rattanaburi S, Poovorawan Y, Zhang Q, Payungporn S. Enhancing the yield of seasonal influenza viruses through manipulation of microRNAs in Madin-Darby canine kidney cells. Exp Biol Med (Maywood) 2022; 247:1335-1349. [PMID: 35666095 PMCID: PMC9442458 DOI: 10.1177/15353702221098340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Annual influenza vaccine is recommended to reduce the occurrence of seasonal influenza and its complications. Thus far, Madin-Darby canine kidney (MDCK) cell line has been used to manufacture cell-based influenza vaccines. Even though host microRNAs may facilitate viral replication, the interaction between MDCK cells-derived microRNAs and seasonal influenza viruses has been less frequently investigated. Therefore, this study highlighted microRNA profiles of MDCK cells to increase the yield of seasonal influenza virus production by manipulating cellular microRNAs. MDCK cells were infected with influenza A or B virus at a multiplicity of infection (MOI) of 0.01, and microRNA collections were then subjected to MiSeq (Illumina) Sequencing. The validated profiles revealed that cfa-miR-340, cfa-miR-146b, cfa-miR-197, and cfa-miR-215 were the most frequently upregulated microRNAs. The effect of candidate microRNA inhibition and overexpression on viral replication was determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). The hybridization pattern between candidate miRNAs and viral genes was performed using miRBase and RNAhybrid web-based programs. Moreover, the predicted microRNA-binding sites were validated by a 3'-UTR reporter assay. The results indicated that cfa-miR-146b could directly target the PB1 gene of A/pH1N1 and the PA gene of B/Yamagata. Furthermore, cfa-miR-215 could silence the PB1 gene of A/pH1N1 and the PB1 gene of B/Victoria. However, the PB2 gene of the A/H3N2 virus was silenced by cfa-miR-197. In addition, the HA and NA sequences of influenza viruses harvested from the cell cultures treated with microRNA inhibitors were analyzed. The sequencing results revealed no difference in the antigenic HA and NA sequences between viruses isolated from the cells treated with microRNA inhibitors and the parental viruses. In conclusion, these findings suggested that MDCK cell-derived microRNAs target viral genes in a strain-specific manner for suppressing viral replication. Conversely, the use of such microRNA inhibitors may facilitate the production of influenza viruses.
Collapse
Affiliation(s)
- Suthat Saengchoowong
- Joint Chulalongkorn
University-University of Liverpool Doctoral Program in Biomedical Sciences and
Biotechnology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330,
Thailand,Faculty of Veterinary Medicine and
Applied Zoology, HRH Princess Chulabhorn College of Medical Science, Chulabhorn
Royal Academy, Bangkok 10210, Thailand
| | - Pattaraporn Nimsamer
- Research Unit of Systems Microbiology,
Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok
10330, Thailand
| | - Kritsada Khongnomnan
- Research Unit of Systems Microbiology,
Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok
10330, Thailand
| | - Witthaya Poomipak
- Research Affairs, Faculty of Medicine,
Chulalongkorn University, Bangkok 10330, Thailand
| | - Kesmanee Praianantathavorn
- Research Unit of Systems Microbiology,
Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok
10330, Thailand
| | - Somruthai Rattanaburi
- Research Unit of Systems Microbiology,
Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok
10330, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical
Virology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330,
Thailand
| | - Qibo Zhang
- Department of Clinical Infection,
Microbiology and Immunology, Institute of Infection, Veterinary and Ecological
Sciences, University of Liverpool, Liverpool L69 7BE, UK
| | - Sunchai Payungporn
- Research Unit of Systems Microbiology,
Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok
10330, Thailand,Sunchai Payungporn.
| |
Collapse
|
5
|
Nogales A, DeDiego ML, Martínez-Sobrido L. Live attenuated influenza A virus vaccines with modified NS1 proteins for veterinary use. Front Cell Infect Microbiol 2022; 12:954811. [PMID: 35937688 PMCID: PMC9354547 DOI: 10.3389/fcimb.2022.954811] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Influenza A viruses (IAV) spread rapidly and can infect a broad range of avian or mammalian species, having a tremendous impact in human and animal health and the global economy. IAV have evolved to develop efficient mechanisms to counteract innate immune responses, the first host mechanism that restricts IAV infection and replication. One key player in this fight against host-induced innate immune responses is the IAV non-structural 1 (NS1) protein that modulates antiviral responses and virus pathogenicity during infection. In the last decades, the implementation of reverse genetics approaches has allowed to modify the viral genome to design recombinant IAV, providing researchers a powerful platform to develop effective vaccine strategies. Among them, different levels of truncation or deletion of the NS1 protein of multiple IAV strains has resulted in attenuated viruses able to induce robust innate and adaptive immune responses, and high levels of protection against wild-type (WT) forms of IAV in multiple animal species and humans. Moreover, this strategy allows the development of novel assays to distinguish between vaccinated and/or infected animals, also known as Differentiating Infected from Vaccinated Animals (DIVA) strategy. In this review, we briefly discuss the potential of NS1 deficient or truncated IAV as safe, immunogenic and protective live-attenuated influenza vaccines (LAIV) to prevent disease caused by this important animal and human pathogen.
Collapse
Affiliation(s)
- Aitor Nogales
- Centro de Investigación en Sanidad Animal (CISA), Centro Nacional Instituto de Investigación y Tecnología Agraria y Alimentaria (INIA, CSIC), Madrid, Spain
- *Correspondence: Aitor Nogales, ; Luis Martínez-Sobrido,
| | - Marta L. DeDiego
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Martínez-Sobrido
- Department of Disease Intervention and Prevetion, Texas Biomedical Research Institute, San Antonio, TX, United States
- *Correspondence: Aitor Nogales, ; Luis Martínez-Sobrido,
| |
Collapse
|
6
|
CRISPR-Cas9 mediated knockout of AnxA6 gene enhances influenza A virus replication in low-permissive HEK293FT cell line. Gene 2022; 809:146024. [PMID: 34673207 DOI: 10.1016/j.gene.2021.146024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/26/2021] [Accepted: 10/14/2021] [Indexed: 11/21/2022]
Abstract
Using cell cultures of human origin for the propagation of influenza virus is an attractive way to preserve its glycosylation profile and antigenic properties, which is essential in influenza surveillance and vaccine production. However, only few cell lines are highly permissive to influenza virus, and none of them are of human origin. The barrier might be associated with host restriction factors inhibiting influenza growth, such as AnxA6 protein counteracting the process of influenza virion packaging. In the presented work we explore the CRISPR-Cas9 mediated knockout of ANXA6 gene as a way to overcome the host restriction barrier and increase the susceptibility of human cell line to influenza infection. By CRISPR-Cas9 genome editing we modified HEK293FT cells and obtained several clones defective in the ANXA6 gene. The replication of the influenza A virus in original HEK293FT cells and the HEK293FT-ANXA6-/- mutant cells was compared in growth curve experiments. By combination of methods including TCID assay and flow cytometry we showed that accumulation of influenza A virus in the mutant HEK293FT-ANXA6-/- cells significantly exceeded the virus titer in the original HEK293FT cells.
Collapse
|
7
|
Dai X, Miao Y, Han P, Zhang X, Yang S, Lv Q, Hua D. PABPC1 Enables Cells with the Suspension Cultivation Feature. ACS Synth Biol 2021; 10:309-317. [PMID: 33502842 DOI: 10.1021/acssynbio.0c00440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cell-based vaccine manufacturing is an important strategy for viral disease prevention. Cultivating cells in suspension could maximize the utility of large bioreactors for cost-effective and scaled up vaccine production, where adapting adherent cells to suspension culture is the bottleneck and key. Through whole transcriptome sequencing of suspension and adherent strains of BHK-21 and CHO-K1 cells followed by the identification of differentially expressed genes, mutational analysis, gene ontology, and pathway enrichment analysis, we identified four candidate genes, PABPC1, LARS, GLUL, PFN1, feasible for genetically modulating anchorage-dependent cells toward cell suspension culture, and experimentally validated the functionality of PABPC1 in both BHK-21 and CHO-K1 cells. Our study unveiled a novel role of PABPC1 that could potentially aid in the establishment of a cost-effective vaccine manufacturing platform relying on cell cultivation in suspension.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Wuhan Ammunition Life-tech Company, Ltd., Wuhan, Hubei 430200, China
| | - Yujie Miao
- School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Peiyu Han
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Xuanhao Zhang
- School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Siming Yang
- School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Qing Lv
- Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| | - Dong Hua
- Affiliated Hospital of Jiangnan University, Wuxi, 214000, China
| |
Collapse
|
8
|
Kiesslich S, Kamen AA. Vero cell upstream bioprocess development for the production of viral vectors and vaccines. Biotechnol Adv 2020; 44:107608. [PMID: 32768520 PMCID: PMC7405825 DOI: 10.1016/j.biotechadv.2020.107608] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022]
Abstract
The Vero cell line is considered the most used continuous cell line for the production of viral vectors and vaccines. Historically, it is the first cell line that was approved by the WHO for the production of human vaccines. Comprehensive experimental data on the production of many viruses using the Vero cell line can be found in the literature. However, the vast majority of these processes is relying on the microcarrier technology. While this system is established for the large-scale manufacturing of viral vaccine, it is still quite complex and labor intensive. Moreover, scale-up remains difficult and is limited by the surface area given by the carriers. To overcome these and other drawbacks and to establish more efficient manufacturing processes, it is a priority to further develop the Vero cell platform by applying novel bioprocess technologies. Especially in times like the current COVID-19 pandemic, advanced and scalable platform technologies could provide more efficient and cost-effective solutions to meet the global vaccine demand. Herein, we review the prevailing literature on Vero cell bioprocess development for the production of viral vectors and vaccines with the aim to assess the recent advances in bioprocess development. We critically underline the need for further research activities and describe bottlenecks to improve the Vero cell platform by taking advantage of recent developments in the cell culture engineering field.
Collapse
Affiliation(s)
- Sascha Kiesslich
- Department of Bioengineering, McGill University, 817 Sherbrooke Street West, Montreal, Quebec H3A 0C3, Canada
| | - Amine A Kamen
- Department of Bioengineering, McGill University, 817 Sherbrooke Street West, Montreal, Quebec H3A 0C3, Canada.
| |
Collapse
|
9
|
Dai X, Zhang X, Ostrikov K, Abrahamyan L. Host receptors: the key to establishing cells with broad viral tropism for vaccine production. Crit Rev Microbiol 2020; 46:147-168. [PMID: 32202955 PMCID: PMC7113910 DOI: 10.1080/1040841x.2020.1735992] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell culture-based vaccine technology is a flexible and convenient approach for vaccine production that requires adaptation of the vaccine strains to the new cells. Driven by the motivation to develop a broadly permissive cell line for infection with a wide range of viruses, we identified a set of the most relevant host receptors involved in viral attachment and entry. This identification was done through a review of different viral entry pathways and host cell lines, and in the context of the Baltimore classification of viruses. In addition, we indicated the potential technical problems and proposed some solutions regarding how to modify the host cell genome in order to meet industrial requirements for mass production of antiviral vaccines. Our work contributes to a finer understanding of the importance of breaking the host–virus recognition specificities for the possibility of creating a cell line feasible for the production of vaccines against a broad spectrum of viruses.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xuanhao Zhang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Kostya Ostrikov
- School of Chemistry and Physics and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Levon Abrahamyan
- Faculty of Veterinary Medicine, Swine and Poultry Infectious Diseases Research Center (CRIPA), Research Group on Infectious Diseases in Production Animals (GREMIP), Université de Montréal, Saint-Hyacinthe, Canada
| |
Collapse
|
10
|
Lee DK, Park J, Seo DW. Suspension culture of Vero cells for the production of adenovirus type 5. Clin Exp Vaccine Res 2020; 9:48-55. [PMID: 32095440 PMCID: PMC7024729 DOI: 10.7774/cevr.2020.9.1.48] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/20/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose Most cell culture processes for viral vaccine production are mainly based on adherent cell culture systems using serum, which are associated with expensive and labor-intensive processes to produce large amounts of viral vaccine strains. In this study, we investigated whether Vero cells could be grown in serum-free and shaking suspension conditions. Furthermore, we assessed the ability of the Vero cell suspension culture system to produce adenovirus type 5 (Ad5), compared to that of the adhesive Vero cell culture system. Materials and Methods We tested the feasibility of commercial serum-free media for Vero cell culture. For the adaptation of Vero cells in suspension culture, adhesive Vero cells were added in the early phase of shaking suspension culture, and 50 days after shaking suspension culture, suspension-adapted Vero cells were subcultured continuously. To assess the virus production ability of Vero cells in suspension, the cells were infected with Ad5-green fluorescent protein and evaluated based on their fluorescence intensity. Results The Vero cells grown in OptiPRO serum-free medium showed no changes in morphology and growth rate, but MRC-5 and FRhk-4 cells showed morphological changes and decreased growth rate, respectively. The Vero cells were well adapted to the suspension culture system. The Vero cells in suspension showed a better Ad5 production ability than the adherent Vero cells. Conclusion Vero cells can be grown in OptiPRO serum-free medium. Further, our suspension culture-adapted Vero cells may be suitable to produce viral vaccine strains due to their high ability to produce viruses such as Ad5.
Collapse
Affiliation(s)
- Deuk-Ki Lee
- Gyeongbuk Institute for Bio Industry, Andong, Korea
| | - Jihye Park
- Gyeongbuk Institute for Bio Industry, Andong, Korea
| | - Dong-Won Seo
- Gyeongbuk Institute for Bio Industry, Andong, Korea
| |
Collapse
|
11
|
Blanco-Lobo P, Nogales A, Rodríguez L, Martínez-Sobrido L. Novel Approaches for The Development of Live Attenuated Influenza Vaccines. Viruses 2019; 11:E190. [PMID: 30813325 PMCID: PMC6409754 DOI: 10.3390/v11020190] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/19/2019] [Accepted: 02/19/2019] [Indexed: 01/04/2023] Open
Abstract
Influenza virus still represents a considerable threat to global public health, despite the advances in the development and wide use of influenza vaccines. Vaccination with traditional inactivate influenza vaccines (IIV) or live-attenuated influenza vaccines (LAIV) remains the main strategy in the control of annual seasonal epidemics, but it does not offer protection against new influenza viruses with pandemic potential, those that have shifted. Moreover, the continual antigenic drift of seasonal circulating influenza viruses, causing an antigenic mismatch that requires yearly reformulation of seasonal influenza vaccines, seriously compromises vaccine efficacy. Therefore, the quick optimization of vaccine production for seasonal influenza and the development of new vaccine approaches for pandemic viruses is still a challenge for the prevention of influenza infections. Moreover, recent reports have questioned the effectiveness of the current LAIV because of limited protection, mainly against the influenza A virus (IAV) component of the vaccine. Although the reasons for the poor protection efficacy of the LAIV have not yet been elucidated, researchers are encouraged to develop new vaccination approaches that overcome the limitations that are associated with the current LAIV. The discovery and implementation of plasmid-based reverse genetics has been a key advance in the rapid generation of recombinant attenuated influenza viruses that can be used for the development of new and most effective LAIV. In this review, we provide an update regarding the progress that has been made during the last five years in the development of new LAIV and the innovative ways that are being explored as alternatives to the currently licensed LAIV. The safety, immunogenicity, and protection efficacy profile of these new LAIVs reveal their possible implementation in combating influenza infections. However, efforts by vaccine companies and government agencies will be needed for controlled testing and approving, respectively, these new vaccine methodologies for the control of influenza infections.
Collapse
Affiliation(s)
- Pilar Blanco-Lobo
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| | - Aitor Nogales
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| | - Laura Rodríguez
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, NY 14642, USA.
| |
Collapse
|
12
|
Pérez Rubio A, Eiros JM. Cell culture-derived flu vaccine: Present and future. Hum Vaccin Immunother 2018; 14:1874-1882. [PMID: 29672213 PMCID: PMC6149758 DOI: 10.1080/21645515.2018.1460297] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/12/2018] [Accepted: 03/29/2018] [Indexed: 12/14/2022] Open
Abstract
The benefit of influenza vaccines is difficult to estimate due to the complexity of accurately assessing the burden of influenza. To improve the efficacy of influenza vaccines, vaccine manufacturers have developed quadrivalent influenza vaccine (QIV) formulations for seasonal vaccination by including both influenza B lineages. Three parallel approaches for producing influenza vaccines are attracting the interest of many vaccine manufacturing companies. The first and oldest is the conventional egg-derived influenza vaccine, which is used by the current licensed influenza vaccines. The second approach is a cell culture-derived influenza vaccine, and the third and most recent is synthetic vaccines. Here, we analyze the difficulties with vaccines production in eggs and compare this to cell culture-derived influenza vaccines and discuss the future of cell culture-derived QIVs.
Collapse
Affiliation(s)
| | - Jose María Eiros
- Servicio Microbiología, Hospital Universitario Rio Hortega, Valladolid, Spain
| |
Collapse
|
13
|
Gonzàlez-Parra G, De Ridder F, Huntjens D, Roymans D, Ispas G, Dobrovolny HM. A comparison of RSV and influenza in vitro kinetic parameters reveals differences in infecting time. PLoS One 2018; 13:e0192645. [PMID: 29420667 PMCID: PMC5805318 DOI: 10.1371/journal.pone.0192645] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 01/26/2018] [Indexed: 11/19/2022] Open
Abstract
Influenza and respiratory syncytial virus (RSV) cause acute infections of the respiratory tract. Since the viruses both cause illnesses with similar symptoms, researchers often try to apply knowledge gleaned from study of one virus to the other virus. This can be an effective and efficient strategy for understanding viral dynamics or developing treatment strategies, but only if we have a full understanding of the similarities and differences between the two viruses. This study used mathematical modeling to quantitatively compare the viral kinetics of in vitro RSV and influenza virus infections. Specifically, we determined the viral kinetics parameters for RSV A2 and three strains of influenza virus, A/WSN/33 (H1N1), A/Puerto Rico/8/1934 (H1N1), and pandemic H1N1 influenza virus. We found that RSV viral titer increases at a slower rate and reaches its peak value later than influenza virus. Our analysis indicated that the slower increase of RSV viral titer is caused by slower spreading of the virus from one cell to another. These results provide estimates of dynamical differences between influenza virus and RSV and help provide insight into the virus-host interactions that cause observed differences in the time courses of the two illnesses in patients.
Collapse
Affiliation(s)
- Gilberto Gonzàlez-Parra
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States of America
- Department of Mathematics, New Mexico Tech, Socorro, NM, United States of America
| | | | | | | | | | - Hana M. Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States of America
- * E-mail:
| |
Collapse
|
14
|
Shen C, Liu Q, Zhou Y, Ku Z, Wang L, Lan K, Ye X, Huang Z. Inactivated coxsackievirus A10 experimental vaccines protect mice against lethal viral challenge. Vaccine 2016; 34:5005-5012. [DOI: 10.1016/j.vaccine.2016.08.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/06/2016] [Accepted: 08/10/2016] [Indexed: 01/02/2023]
|
15
|
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: 50] [Impact Index Per Article: 5.6] [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
|
16
|
Krömmelbein N, Wiebusch L, Schiedner G, Büscher N, Sauer C, Florin L, Sehn E, Wolfrum U, Plachter B. Adenovirus E1A/E1B Transformed Amniotic Fluid Cells Support Human Cytomegalovirus Replication. Viruses 2016; 8:v8020037. [PMID: 26848680 PMCID: PMC4776192 DOI: 10.3390/v8020037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/22/2016] [Accepted: 01/28/2016] [Indexed: 11/30/2022] Open
Abstract
The human cytomegalovirus (HCMV) replicates to high titers in primary human fibroblast cell cultures. A variety of primary human cells and some tumor-derived cell lines do also support permissive HCMV replication, yet at low levels. Cell lines established by transfection of the transforming functions of adenoviruses have been notoriously resistant to HCMV replication and progeny production. Here, we provide first-time evidence that a permanent cell line immortalized by adenovirus type 5 E1A and E1B (CAP) is supporting the full HCMV replication cycle and is releasing infectious progeny. The CAP cell line had previously been established from amniotic fluid cells which were likely derived from membranes of the developing fetus. These cells can be grown under serum-free conditions. HCMV efficiently penetrated CAP cells, expressed its immediate-early proteins and dispersed restrictive PML-bodies. Viral DNA replication was initiated and viral progeny became detectable by electron microscopy in CAP cells. Furthermore, infectious virus was released from CAP cells, yet to lower levels compared to fibroblasts. Subviral dense bodies were also secreted from CAP cells. The results show that E1A/E1B expression in transformed cells is not generally repressive to HCMV replication and that CAP cells may be a good substrate for dense body based vaccine production.
Collapse
Affiliation(s)
- Natascha Krömmelbein
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Lüder Wiebusch
- Department of Pediatric Molecular Biology, Charité University Medical Centre Berlin, D-10117 Berlin, Germany.
| | | | - Nicole Büscher
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Caroline Sauer
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Luise Florin
- Institute for Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| | - Elisabeth Sehn
- Institute for Zoology, Johannes Gutenberg-University Mainz, D-55099 Mainz, Germany.
| | - Uwe Wolfrum
- Institute for Zoology, Johannes Gutenberg-University Mainz, D-55099 Mainz, Germany.
| | - Bodo Plachter
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany.
| |
Collapse
|
17
|
Li J, Liu G, Liu X, Yang J, Chang J, Zhang W, Yu XF. Optimization and Characterization of Candidate Strain for Coxsackievirus A16 Inactivated Vaccine. Viruses 2015; 7:3891-909. [PMID: 26193302 PMCID: PMC4517132 DOI: 10.3390/v7072803] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/20/2015] [Accepted: 07/01/2015] [Indexed: 11/16/2022] Open
Abstract
Coxsackievirus A16 (CA16) and enterovirus 71 (EV71), both of which can cause hand, foot and mouth disease (HFMD), are responsible for large epidemics in Asian and Pacific areas. Although inactivated EV71 vaccines have completed testing in phase III clinical trials in Mainland China, CA16 vaccines are still under development. A Vero cell-based inactivated CA16 vaccine was developed by our group. Screening identified a CA16 vaccine strain (CC024) isolated from HFMD patients, which had broad cross-protective abilities and satisfied all requirements for vaccine production. Identification of the biological characteristics showed that the CA16CC024 strain had the highest titer (107.5 CCID50/mL) in Vero cells, which would benefit the development of an EV71/CA16 divalent vaccine. A potential vaccine manufacturing process was established, including the selection of optimal time for virus harvesting, membrane for diafiltration and concentration, gel-filtration chromatography for the down-stream virus purification and virus inactivation method. Altogether, the analyses suggested that the CC-16, a limiting dilution clone of the CC024 strain, with good genetic stability, high titer and broad-spectrum immunogenicity, would be the best candidate strain for a CA16 inactivated vaccine. Therefore, our study provides valuable information for the development of a Vero cell-based CA16 or EV71-CA16 divalent inactivated vaccine.
Collapse
Affiliation(s)
- Jingliang Li
- First Hospital of Jilin University, Institute of Virology and AIDS Research, 130061 Changchun, China.
| | - Guanchen Liu
- First Hospital of Jilin University, Institute of Virology and AIDS Research, 130061 Changchun, China.
| | - Xin Liu
- First Hospital of Jilin University, Institute of Virology and AIDS Research, 130061 Changchun, China.
| | - Jiaxin Yang
- First Hospital of Jilin University, Institute of Virology and AIDS Research, 130061 Changchun, China.
| | - Junliang Chang
- First Hospital of Jilin University, Institute of Virology and AIDS Research, 130061 Changchun, China.
| | - Wenyan Zhang
- First Hospital of Jilin University, Institute of Virology and AIDS Research, 130061 Changchun, China.
| | - Xiao-Fang Yu
- First Hospital of Jilin University, Institute of Virology and AIDS Research, 130061 Changchun, China.
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
18
|
Gallo-Ramírez LE, Nikolay A, Genzel Y, Reichl U. Bioreactor concepts for cell culture-based viral vaccine production. Expert Rev Vaccines 2015; 14:1181-95. [PMID: 26178380 DOI: 10.1586/14760584.2015.1067144] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Vaccine manufacturing processes are designed to meet present and upcoming challenges associated with a growing vaccine market and to include multi-use facilities offering a broad portfolio and faster reaction times in case of pandemics and emerging diseases. The final products, from whole viruses to recombinant viral proteins, are very diverse, making standard process strategies hardly universally applicable. Numerous factors such as cell substrate, virus strain or expression system, medium, cultivation system, cultivation method, and scale need consideration. Reviewing options for efficient and economical production of human vaccines, this paper discusses basic factors relevant for viral antigen production in mammalian cells, avian cells and insect cells. In addition, bioreactor concepts, including static systems, single-use systems, stirred tanks and packed-beds are addressed. On this basis, methods towards process intensification, in particular operational strategies, the use of perfusion systems for high product yields, and steps to establish continuous processes are introduced.
Collapse
Affiliation(s)
- Lilí Esmeralda Gallo-Ramírez
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg; Sandtorstr. 1, 39106 Magdeburg, Germany
| | | | | | | |
Collapse
|
19
|
Carvajal-Yepes M, Sporer KRB, Carter JL, Colvin CJ, Coussens PM. Enhanced production of human influenza virus in PBS-12SF cells with a reduced interferon response. Hum Vaccin Immunother 2015; 11:2296-304. [PMID: 26090991 DOI: 10.1080/21645515.2015.1016677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Influenza is one of the most important infectious diseases in humans. The best way to prevent severe illness caused by influenza infection is vaccination. Cell culture-derived influenza vaccines are being considered in addition to the widely used egg-based system in order to support the increasing seasonal demand and to be prepared in case of a pandemic. Cell culture based systems offer increased safety, capacity, and flexibility with reduced downstream processing relative to embryonated eggs. We have previously reported a chick embryo cell line, termed PBS-12SF, that supports replication of human and avian influenza A viruses to high titers (>10(7) PFU/ml) without the need for exogenous proteases or serum proteins. Viral infections in cells are limited by the Interferon (IFN) response typified by production of type I IFNs that bind to the IFNα/β receptor and activate an antiviral state. In this study, we investigated how neutralizing the interferon (IFN) response in PBS-12SF cells, via shRNA-mediated knock-down of IFNAR1 mRNA expression, affects influenza virus production. We were successful in knocking down ∼90% of IFNAR1 protein expression by this method, resulting in a significant decrease in the response to recombinant chIFNα stimulation in PBS-12SF cells as shown by a reduction in expression of interferon-responsive genes when compared to control cells. Additionally; IFNAR1-knock-down cells displayed enhanced viral HA production and released more virus into cell culture supernatants than parental PBS-12SF cells.
Collapse
Affiliation(s)
- Monica Carvajal-Yepes
- a Molecular Pathogenesis Laboratory; Department of Animal Science; Michigan State University ; East Lansing , MI USA
| | | | | | | | | |
Collapse
|
20
|
Genzel Y. Designing cell lines for viral vaccine production: Where do we stand? Biotechnol J 2015; 10:728-40. [PMID: 25903999 DOI: 10.1002/biot.201400388] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 03/24/2015] [Accepted: 03/31/2015] [Indexed: 12/11/2022]
Abstract
Established animal cells, such as Vero, Madin Darby canine kidney (MDCK) or chicken embryo fibroblasts (CEFs), are still the main cell lines used for viral vaccine production, although new "designer cells" have been available for some years. These designer cell lines were specifically developed as a cell substrate for one application and are well characterized. Later screening for other possible applications widened the product range. These cells grow in suspension in chemically defined media under controlled conditions and can be used for up to 100 passages. Scale-up is easier and current process options allow cultivation in disposable bioreactors at cell concentrations higher than 1 × 10(7) cells/mL. This review covers the limitations of established cell lines and discusses the requirements and screening options for new host cells. Currently available designer cells for viral vaccine production (PER.C6, CAP, AGE1.CR, EB66 cells), together with other new cell lines (PBS-1, QOR/2E11, SogE, MFF-8C1 cells) that were recently described as possible cell substrates are presented. Using current process knowledge and cell line development tools, future upstream processing could resemble today's Chinese hamster ovary (CHO) cell processes for monoclonal antibody production: small scale bioreactors (disposable) in perfusion or fed-batch mode with cell concentrations above 1 × 10(8) cells/mL.
Collapse
Affiliation(s)
- Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.
| |
Collapse
|
21
|
Hematopoietic cancer cell lines can support replication of Sabin poliovirus type 1. BIOMED RESEARCH INTERNATIONAL 2015; 2015:358462. [PMID: 25815312 PMCID: PMC4359862 DOI: 10.1155/2015/358462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 02/17/2015] [Accepted: 02/17/2015] [Indexed: 02/02/2023]
Abstract
Viral vaccines can be produced in adherent or in suspension cells. The objective of this work was to screen human suspension cell lines for the capacity to support viral replication. As the first step, it was investigated whether poliovirus can replicate in such cell lines. Sabin poliovirus type 1 was serially passaged on five human cell lines, HL60, K562, KG1, THP-1, and U937. Sabin type 1 was capable of efficiently replicating in three cell lines (K562, KG1, and U937), yielding high viral titers after replication. Expression of CD155, the poliovirus receptor, did not explain susceptibility to replication, since all cell lines expressed CD155. Furthermore, we showed that passaged virus replicated more efficiently than parental virus in KG1 cells, yielding higher virus titers in the supernatant early after infection. Infection of cell lines at an MOI of 0.01 resulted in high viral titers in the supernatant at day 4. Infection of K562 with passaged Sabin type 1 in a bioreactor system yielded high viral titers in the supernatant. Altogether, these data suggest that K562, KG1, and U937 cell lines are useful for propagation of poliovirus.
Collapse
|
22
|
Medina J, Guillot V, Totain E, Rouleau M, Sodoyer R, Moste C, Legastelois I. Vero/CHOK1, a novel mixture of cell lines that is optimal for the rescue of influenza A vaccine seeds. J Virol Methods 2014; 196:25-31. [DOI: 10.1016/j.jviromet.2013.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 06/12/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
|
23
|
Guo D, Zhu Q, Zhang H, Sun D. Proteomic analysis of membrane proteins of vero cells: exploration of potential proteins responsible for virus entry. DNA Cell Biol 2013; 33:20-8. [PMID: 24286161 DOI: 10.1089/dna.2013.2193] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vero cells are highly susceptible to many viruses in humans and animals, and its membrane proteins (MPs) are responsible for virus entry. In our study, the MP proteome of the Vero cells was investigated using a shotgun LC-MS/MS approach. Six hundred twenty-seven proteins, including a total of 1839 peptides, were identified in MP samples of the Vero cells. In 627 proteins, 307 proteins (48.96%) were annotated in terms of biological process of gene ontology (GO) categories; 356 proteins (56.78%) were annotated in terms of molecular function of GO categories; 414 proteins (66.03%) were annotated in terms of cellular components of GO categories. Of 627 identified proteins, seventeen proteins had been revealed to be virus receptor proteins. The resulting protein lists and highlighted proteins may provide valuable information to increase understanding of virus infection of Vero cells.
Collapse
Affiliation(s)
- Donghua Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University , Daqing, People's Republic of China
| | | | | | | |
Collapse
|