1
|
Silva CAT, Kamen AA, Henry O. Fed-batch strategies for intensified rVSV vector production in high cell density cultures of suspension HEK293 cells. Biotechnol Prog 2025; 41:e3506. [PMID: 39286892 PMCID: PMC11831413 DOI: 10.1002/btpr.3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
Vesicular stomatitis virus (VSV) has been increasingly demonstrated as a promising viral vector platform. As the interest over this modality for vaccine and gene therapy applications increases, the need for intensified processes to produce these vectors emerge. In this study, we develop fed-batch-based operations to intensify the production of a recombinant VSV-based vaccine candidate (rVSV-SARS-CoV-2) in suspension cultures of HEK293 cells. A feeding strategy, in which a commercial concentrated medium was added to cultures based on cell growth through a fixed cell specific feeding rate (CSFR), was applied for the development of two different processes using Ambr250 modular bioreactors. Cultures operated in hybrid fed-batch/perfusion (FB/P) or fed-batch (FB) were able to sustain infections performed at 8.0 × 106 cells/mL, respectively resulting in 3.9 and 5.0-fold increase in total yield (YT) and 1.7 and 5.6-fold increase in volumetric productivity (VP) when compared with a batch reference. A maximum viral titer of 4.5 × 1010 TCID50/mL was reached, which is comparable or higher than other processes for VSV production in different cell lines. Overall, our study reports efficient fed-batch options to intensify the production of a rVSV-based vaccine candidate in suspension HEK293 cells.
Collapse
Affiliation(s)
- Cristina A. T. Silva
- Department of Chemical EngineeringPolytechnique MontréalMontrealQuebecCanada
- Department of BioengineeringMcGill UniversityMontrealQuebecCanada
| | - Amine A. Kamen
- Department of BioengineeringMcGill UniversityMontrealQuebecCanada
| | - Olivier Henry
- Department of Chemical EngineeringPolytechnique MontréalMontrealQuebecCanada
| |
Collapse
|
2
|
Göbel S, Kazemi O, Ma J, Jordan I, Sandig V, Paulissen J, Kerstens W, Thibaut HJ, Reichl U, Dallmeier K, Genzel Y. Parallel Multifactorial Process Optimization and Intensification for High-Yield Production of Live YF17D-Vectored Zika Vaccine. Vaccines (Basel) 2024; 12:755. [PMID: 39066393 PMCID: PMC11281342 DOI: 10.3390/vaccines12070755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
The live-attenuated yellow fever 17D strain is a potent vaccine and viral vector. Its manufacture is based on embryonated chicken eggs or adherent Vero cells. Both processes are unsuitable for rapid and scalable supply. Here, we introduce a high-throughput workflow to identify suspension cells that are fit for the high-yield production of live YF17D-based vaccines in an intensified upstream process. The use of an automated parallel ambr15 microbioreactor system for screening and process optimization has led to the identification of two promising cell lines (AGE1.CR.pIX and HEKDyn) and the establishment of optimized production conditions, which have resulted in a >100-fold increase in virus titers compared to the current state of the art using adherent Vero cells. The process can readily be scaled up from the microbioreactor scale (15 mL) to 1 L stirred tank bioreactors. The viruses produced are genetically stable and maintain their favorable safety and immunogenicity profile, as demonstrated by the absence of neurovirulence in suckling BALB/c mice and consistent seroprotection in AG129 mice. In conclusion, the presented workflow allows for the rapid establishment of a robust, scalable, and high-yield process for the production of live-attenuated orthoflavivirus vaccines, which outperforms current standards. The approach described here can serve as a model for the development of scalable processes and the optimization of yields for other virus-based vaccines that face challenges in meeting growing demands.
Collapse
Affiliation(s)
- Sven Göbel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
| | - Ozeir Kazemi
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium; (K.D.)
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium; (K.D.)
| | | | | | - Jasmine Paulissen
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Winnie Kerstens
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
- Bioprocess Engineering, Otto-von-Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium; (K.D.)
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
| |
Collapse
|
3
|
Coplan L, Zhang Z, Ragone N, Reeves J, Rodriguez A, Shevade A, Bak H, Tustian AD. High-yield recombinant adeno-associated viral vector production by multivariate optimization of bioprocess and transfection conditions. Biotechnol Prog 2024; 40:e3445. [PMID: 38450973 DOI: 10.1002/btpr.3445] [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: 10/17/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 03/08/2024]
Abstract
Recombinant adeno-associated viral vectors (rAAVs) are one of the most used vehicles for gene therapy, with five rAAV therapeutics commercially approved by the FDA. To improve product yield, we optimized the suspension production process of rAAV8 vectors carrying a proprietary transgene using a commercially available transfection reagent, FectoVIR-AAV. Using a miniaturized automated 250 mL scale bioreactor system, we generated models of vector genome (vg) titer, capsid (cp) titer, and Vg:Cp percentage from two multivariate design of experiment studies, one centered around bioreactor operating parameters, and another based on the transfection conditions. Using the optimized process returned from these models, the vector genome titer from the bioreactor was improved to beyond 1 × 1012 vg/mL. Five critical parameters were identified that had large effects on the pre-purification vector quantity-the transfection pH, production pH, complexation time, viable cell density at transfection, and transfection reagent to DNA ratio. The optimized process was further assessed for its performance extending to six AAV serotypes, namely AAV1, AAV2, AAV5, AAV6, AAV8, and AAV9 carrying a transgene encoding for green fluorescent protein (GFP). Five of the six serotypes returned higher vector genome titers than the control condition. These data suggest that the choice of transfection reagent is a major factor in improving vector yield. The multivariate design of experiment approach is a powerful way to optimize production processes, and the optimized process from one AAV vector can to some extent be generalized to other serotypes and transgenes to accelerate development timelines of new programs.
Collapse
Affiliation(s)
- Louis Coplan
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland, USA
| | - Zhe Zhang
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Nicole Ragone
- Research Operations, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - John Reeves
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Audrey Rodriguez
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Aishwarya Shevade
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Hanne Bak
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Andrew D Tustian
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| |
Collapse
|
4
|
Todesco HM, Gafuik C, John CM, Roberts EL, Borys BS, Pawluk A, Kallos MS, Potts KG, Mahoney DJ. High-titer manufacturing of SARS-CoV-2 Spike-pseudotyped VSV in stirred-tank bioreactors. Mol Ther Methods Clin Dev 2024; 32:101189. [PMID: 38327804 PMCID: PMC10847022 DOI: 10.1016/j.omtm.2024.101189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic highlighted the importance of vaccine innovation in public health. Hundreds of vaccines built on numerous technology platforms have been rapidly developed against SARS-CoV-2 since 2020. Like all vaccine platforms, an important bottleneck to viral-vectored vaccine development is manufacturing. Here, we describe a scalable manufacturing protocol for replication-competent SARS-CoV-2 Spike-pseudotyped vesicular stomatitis virus (S-VSV)-vectored vaccines using Vero cells grown on microcarriers in a stirred-tank bioreactor. Using Cytodex 1 microcarriers over 6 days of fed-batch culture, Vero cells grew to a density of 3.95 ± 0.42 ×106 cells/mL in 1-L stirred-tank bioreactors. Ancestral strain S-VSV reached a peak titer of 2.05 ± 0.58 ×108 plaque-forming units (PFUs)/mL at 3 days postinfection. When compared to growth in plate-based cultures, this was a 29-fold increase in virus production, meaning a 1-L bioreactor produces the same amount of virus as 1,284 plates of 15 cm. In addition, the omicron BA.1 S-VSV reached a peak titer of 5.58 ± 0.35 × 106 PFU/mL. Quality control testing showed plate- and bioreactor-produced S-VSV had similar particle-to-PFU ratios and elicited comparable levels of neutralizing antibodies in immunized hamsters. This method should enhance preclinical and clinical development of pseudotyped VSV-vectored vaccines in future pandemics.
Collapse
Affiliation(s)
- Hayley M. Todesco
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Chris Gafuik
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Cini M. John
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Erin L. Roberts
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Breanna S. Borys
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Alexis Pawluk
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Michael S. Kallos
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Kyle G. Potts
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Douglas J. Mahoney
- Arnie Charbonneau Cancer Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Disease, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
5
|
Göbel S, Pelz L, Silva CAT, Brühlmann B, Hill C, Altomonte J, Kamen A, Reichl U, Genzel Y. Production of recombinant vesicular stomatitis virus-based vectors by tangential flow depth filtration. Appl Microbiol Biotechnol 2024; 108:240. [PMID: 38413399 PMCID: PMC10899354 DOI: 10.1007/s00253-024-13078-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
Cell culture-based production of vector-based vaccines and virotherapeutics is of increasing interest. The vectors used not only retain their ability to infect cells but also induce robust immune responses. Using two recombinant vesicular stomatitis virus (rVSV)-based constructs, we performed a proof-of-concept study regarding an integrated closed single-use perfusion system that allows continuous virus harvesting and clarification. Using suspension BHK-21 cells and a fusogenic oncolytic hybrid of vesicular stomatitis virus and Newcastle disease virus (rVSV-NDV), a modified alternating tangential flow device (mATF) or tangential flow depth filtration (TFDF) systems were used for cell retention. As the hollow fibers of the former are characterized by a large internal lumen (0.75 mm; pore size 0.65 μm), membrane blocking by the multi-nucleated syncytia formed during infection could be prevented. However, virus particles were completely retained. In contrast, the TFDF filter unit (lumen 3.15 mm, pore size 2-5 μm) allowed not only to achieve high viable cell concentrations (VCC, 16.4-20.6×106 cells/mL) but also continuous vector harvesting and clarification. Compared to an optimized batch process, 11-fold higher infectious virus titers were obtained in the clarified permeate (maximum 7.5×109 TCID50/mL). Using HEK293-SF cells and a rVSV vector expressing a green fluorescent protein, perfusion cultivations resulted in a maximum VCC of 11.3×106 cells/mL and infectious virus titers up to 7.1×1010 TCID50/mL in the permeate. Not only continuous harvesting but also clarification was possible. Although the cell-specific virus yield decreased relative to a batch process established as a control, an increased space-time yield was obtained. KEY POINTS: • Viral vector production using a TFDF perfusion system resulted in a 460% increase in space-time yield • Use of a TFDF system allowed continuous virus harvesting and clarification • TFDF perfusion system has great potential towards the establishment of an intensified vector production.
Collapse
Affiliation(s)
- Sven Göbel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Lars Pelz
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Cristina A T Silva
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Québec, Canada
| | | | | | - Jennifer Altomonte
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montréal, Québec, Canada
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany
- Chair for Bioprocess Engineering, Otto von Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106, Magdeburg, Germany.
| |
Collapse
|
6
|
Zheng YY, Hu ZN, Zhou GH. A review: analysis of technical challenges in cultured meat production and its commercialization. Crit Rev Food Sci Nutr 2024; 65:1911-1928. [PMID: 38384235 DOI: 10.1080/10408398.2024.2315447] [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] [Indexed: 02/23/2024]
Abstract
The cultured meat technology has developed rapidly in recent years, but there are still many technical challenges that hinder the large-scale production and commercialization of cultured meat. Firstly, it is necessary to lay the foundation for cultured meat production by obtaining seed cells and maintaining stable cell functions. Next, technologies such as bioreactors are used to expand the scale of cell culture, and three-dimensional culture technologies such as scaffold culture or 3D printing are used to construct the three-dimensional structure of cultured meat. At the same time, it can reduce production costs by developing serum-free medium suitable for cultured meat. Finally, the edible quality of cultured meat is improved by evaluating food safety and sensory flavor, and combining ethical and consumer acceptability issues. Therefore, this review fully demonstrates the current development status and existing technical challenges of the cultured meat production technology with regard to the key points described above, in order to provide research ideas for the industrial production of cultured meat.
Collapse
Affiliation(s)
- Yan-Yan Zheng
- College of Food Science and Technology, Nanjing Agricultural University, National Center of Meat Quality and Safety Nanjing, MOST, Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Nanjing, P.R. China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ze-Nan Hu
- College of Food Science and Technology, Nanjing Agricultural University, National Center of Meat Quality and Safety Nanjing, MOST, Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Nanjing, P.R. China
| | - Guang-Hong Zhou
- College of Food Science and Technology, Nanjing Agricultural University, National Center of Meat Quality and Safety Nanjing, MOST, Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Nanjing, P.R. China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
7
|
Cohen N, Simon I, Hazan O, Tal A, Tzadok H, Levin L, Girshengorn M, Mimran LC, Natan N, Baruhi T, David AB, Rosen O, Shmaya S, Borni S, Cohen N, Lupu E, Kedmi A, Zilberman O, Jayson A, Monash A, Dor E, Diamant E, Goldvaser M, Cohen-Gihon I, Israeli O, Lazar S, Shifman O, Beth-Din A, Zvi A, Oren Z, Makovitzki A, Lerer E, Mimran A, Toister E, Zichel R, Adar Y, Epstein E. Enhanced production yields of rVSV-SARS-CoV-2 vaccine using Fibra-Cel ® macrocarriers. Front Bioeng Biotechnol 2024; 12:1333548. [PMID: 38449674 PMCID: PMC10915211 DOI: 10.3389/fbioe.2024.1333548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/02/2024] [Indexed: 03/08/2024] Open
Abstract
The COVID-19 pandemic has led to high global demand for vaccines to safeguard public health. To that end, our institute has developed a recombinant viral vector vaccine utilizing a modified vesicular stomatitis virus (VSV) construct, wherein the G protein of VSV is replaced with the spike protein of SARS-CoV-2 (rVSV-ΔG-spike). Previous studies have demonstrated the production of a VSV-based vaccine in Vero cells adsorbed on Cytodex 1 microcarriers or in suspension. However, the titers were limited by both the carrier surface area and shear forces. Here, we describe the development of a bioprocess for rVSV-ΔG-spike production in serum-free Vero cells using porous Fibra-Cel® macrocarriers in fixed-bed BioBLU®320 5p bioreactors, leading to high-end titers. We identified core factors that significantly improved virus production, such as the kinetics of virus production, the use of macrospargers for oxygen supply, and medium replenishment. Implementing these parameters, among others, in a series of GMP production processes improved the titer yields by at least two orders of magnitude (2e9 PFU/mL) over previously reported values. The developed process was highly effective, repeatable, and robust, creating potent and genetically stable vaccine viruses and introducing new opportunities for application in other viral vaccine platforms.
Collapse
Affiliation(s)
- Noam Cohen
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Irit Simon
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ophir Hazan
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Arnon Tal
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Hanan Tzadok
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Lilach Levin
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Meni Girshengorn
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Lilach Cherry Mimran
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Niva Natan
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Tzadok Baruhi
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Alon Ben David
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Osnat Rosen
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shlomo Shmaya
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Sarah Borni
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Noa Cohen
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Edith Lupu
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Adi Kedmi
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Orian Zilberman
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Avital Jayson
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Arik Monash
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Eyal Dor
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Eran Diamant
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Michael Goldvaser
- Department of Organic Chemistry, Israel Institute for Biological, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Inbar Cohen-Gihon
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ofir Israeli
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shirley Lazar
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ohad Shifman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Adi Beth-Din
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Anat Zvi
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ziv Oren
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Arik Makovitzki
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Elad Lerer
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Avishai Mimran
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Einat Toister
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ran Zichel
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Yaakov Adar
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Eyal Epstein
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| |
Collapse
|
8
|
Ouyang MJ, Ao Z, Olukitibi TA, Lawrynuik P, Shieh C, Kung SKP, Fowke KR, Kobasa D, Yao X. Oral Immunization with rVSV Bivalent Vaccine Elicits Protective Immune Responses, Including ADCC, against Both SARS-CoV-2 and Influenza A Viruses. Vaccines (Basel) 2023; 11:1404. [PMID: 37766083 PMCID: PMC10534613 DOI: 10.3390/vaccines11091404] [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: 07/26/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
COVID-19 and influenza both cause enormous disease burdens, and vaccines are the primary measures for their control. Since these viral diseases are transmitted through the mucosal surface of the respiratory tract, developing an effective and convenient mucosal vaccine should be a high priority. We previously reported a recombinant vesicular stomatitis virus (rVSV)-based bivalent vaccine (v-EM2/SPΔC1Delta) that protects animals from both SARS-CoV-2 and influenza viruses via intramuscular and intranasal immunization. Here, we further investigated the immune response induced by oral immunization with this vaccine and its protective efficacy in mice. The results demonstrated that the oral delivery, like the intranasal route, elicited strong and protective systemic immune responses against SARS-CoV-2 and influenza A virus. This included high levels of neutralizing antibodies (NAbs) against SARS-CoV-2, as well as strong anti-SARS-CoV-2 spike protein (SP) antibody-dependent cellular cytotoxicity (ADCC) and anti-influenza M2 ADCC responses in mice sera. Furthermore, it provided efficient protection against challenge with influenza H1N1 virus in a mouse model, with a 100% survival rate and a significantly low lung viral load of influenza virus. All these findings provide substantial evidence for the effectiveness of oral immunization with the rVSV bivalent vaccine.
Collapse
Affiliation(s)
- Maggie Jing Ouyang
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Titus A. Olukitibi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Peter Lawrynuik
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Christopher Shieh
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
| | - Sam K. P. Kung
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W3, Canada;
| | - Keith R. Fowke
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| | - Darwyn Kobasa
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3L5, Canada
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 508-745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (M.J.O.); (Z.A.); (T.A.O.); (P.L.); (C.S.)
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada; (K.R.F.); (D.K.)
| |
Collapse
|
9
|
Bakhshizadeh Gashti A, Chahal PS, Gaillet B, Garnier A. Purification of recombinant vesicular stomatitis virus-based HIV vaccine candidate. Vaccine 2023; 41:2198-2207. [PMID: 36842887 DOI: 10.1016/j.vaccine.2023.02.058] [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: 10/10/2022] [Revised: 02/10/2023] [Accepted: 02/18/2023] [Indexed: 02/26/2023]
Abstract
In this work, laboratory- and large-scale methods were tested for purification of a human immunodeficiency virus (HIV) vaccine candidate, based on recombinant vesicular stomatitis virus (rVSV). First step of the purification, the clarification of the rVSVs produced in serum-free cell culture medium, was tested by centrifugation and filtration using different filtration media and pore sizes (0.45 to 30 µm). To reduce the supernatant volume and process time, the clarified sample was concentrated by ultrafiltration either using tangential flow filtration or centrifugal-based filtration units, depending on the process scale. The final purification step at laboratory-scale, was carried out by density gradient ultracentrifugation, the recovery of which was compared with chromatographic purification at large-scale. The virus preparations were analyzed using dynamic light scattering to verify the virus size and transmission electron microscopy for purity and virus morphology. Density gradient ultracentrifugation allowed the recovery of ≥ 80% infectious particles and reduced the contaminant DNA and host cell proteins relatively to standard ultracentrifugation pelleting using a sucrose cushion. At large-scale, weak and strong anion-exchangers were tested and compared. The best columns allowed infectious virus recoveries as high as 77% and eliminated 92% of host cell proteins.
Collapse
Affiliation(s)
- Anahita Bakhshizadeh Gashti
- Department of Chemical Engineering, Faculty of Sciences and Engineering, Université Laval, Quebec, QC, Canada; Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Parminder S Chahal
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Bruno Gaillet
- Department of Chemical Engineering, Faculty of Sciences and Engineering, Université Laval, Quebec, QC, Canada
| | - Alain Garnier
- Department of Chemical Engineering, Faculty of Sciences and Engineering, Université Laval, Quebec, QC, Canada.
| |
Collapse
|
10
|
Ton C, Stabile V, Carey E, Maraikar A, Whitmer T, Marrone S, Afanador NL, Zabrodin I, Manomohan G, Whiteman M, Hofmann C. Development and scale-up of rVSV-SARS-CoV-2 vaccine process using single use bioreactor. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2023; 37:e00782. [PMID: 36687766 PMCID: PMC9841742 DOI: 10.1016/j.btre.2023.e00782] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023]
Abstract
The outbreak of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes the Coronavirus Disease 2019 (COVID-19) has spread through the globe at an alarming speed. The disease has become a global pandemic affecting millions of people and created public health crises worldwide. Among many efforts to urgently develop a vaccine against this disease, we developed an industrial-scale closed, single use manufacturing process for V590, a vaccine candidate for SARS-CoV-2. V590 is a recombinant vesicular stomatitis virus (rVSV) genetically engineered to express SARS-CoV-2 glycoprotein. In this work, we describe the development and optimization of serum-free microcarrier production of V590 in Vero cells in a closed system. To achieve the maximum virus productivity, we optimized pH and temperature during virus production in 3 liters (L) bioreactors. Virus productivity was improved (by ∼1 log) by using pH 7.0 and temperature at 34.0 °C. The optimal production condition was successfully scaled up to a 2000 L Single Use Bioreactor (SUB), producing a maximum virus titer of ∼1.0e+7 plaque forming units (PFU)/mL. Further process intensification and simplification, including growing Vero cells at 2 gs per liter (g/L) of Cytodex-1 Gamma microcarriers and eliminating the media exchange (MX) step prior to infection helped to increase virus productivity by ∼2-fold.
Collapse
Affiliation(s)
- Christopher Ton
- Vaccine Process Development, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States,Corresponding author.
| | - Victoria Stabile
- Vaccine Process Development, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States
| | - Elizabeth Carey
- Vaccine Process Development, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States
| | - Adam Maraikar
- Bioprocess Clinical Manufacturing & Technology, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States
| | - Travis Whitmer
- Bioprocess Drug Substance Commercialization, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States
| | - Samantha Marrone
- Vaccine Process Development, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States
| | - Nelson Lee Afanador
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States
| | - Igor Zabrodin
- Vaccine Process Development, Merck & Co., Inc., West Point, Pennsylvania, 19486, United States
| | - Greeshma Manomohan
- Currently at GlaxoSmithKline plc, King of Prussia, Pennsylvania, 19406, United States
| | - Melissa Whiteman
- Analytical Research & Development, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Carl Hofmann
- Analytical Research & Development, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
11
|
Xu WJ, Lin Y, Mi CL, Pang JY, Wang TY. Progress in fed-batch culture for recombinant protein production in CHO cells. Appl Microbiol Biotechnol 2023; 107:1063-1075. [PMID: 36648523 PMCID: PMC9843118 DOI: 10.1007/s00253-022-12342-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023]
Abstract
Nearly 80% of the approved human therapeutic antibodies are produced by Chinese Hamster Ovary (CHO) cells. To achieve better cell growth and high-yield recombinant protein, fed-batch culture is typically used for recombinant protein production in CHO cells. According to the demand of nutrients consumption, feed medium containing multiple components in cell culture can affect the characteristics of cell growth and improve the yield and quality of recombinant protein. Fed-batch optimization should have a connection with comprehensive factors such as culture environmental parameters, feed composition, and feeding strategy. At present, process intensification (PI) is explored to maintain production flexible and meet forthcoming demands of biotherapeutics process. Here, CHO cell culture, feed composition in fed-batch culture, fed-batch culture environmental parameters, feeding strategies, metabolic byproducts in fed-batch culture, chemostat cultivation, and the intensified fed-batch are reviewed. KEY POINTS: • Fed-batch culture in CHO cells is reviewed. • Fed-batch has become a common technology for recombinant protein production. • Fed batch culture promotes recombinant protein production in CHO cells.
Collapse
Affiliation(s)
- Wen-Jing Xu
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China ,grid.412990.70000 0004 1808 322XSchool of Pharmacy, Xinxiang Medical University, Xinxiang, 453003 Henan China
| | - Yan Lin
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China ,grid.412990.70000 0004 1808 322XSchool of Nursing, Xinxiang Medical University, Xinxiang, 453003 Henan China
| | - Chun-Liu Mi
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China
| | - Jing-Ying Pang
- grid.412990.70000 0004 1808 322XSchool of the First Clinical College, Xinxiang Medical University, Xinxiang, 453000 Henan China
| | - Tian-Yun Wang
- grid.412990.70000 0004 1808 322XInternational Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003 Henan China ,grid.495434.b0000 0004 1797 4346School of medicine, Xinxiang University, Xinxiang, 453003 Henan China
| |
Collapse
|
12
|
Fang Z, Lyu J, Li J, Li C, Zhang Y, Guo Y, Wang Y, Zhang Y, Chen K. Application of bioreactor technology for cell culture-based viral vaccine production: Present status and future prospects. Front Bioeng Biotechnol 2022; 10:921755. [PMID: 36017347 PMCID: PMC9395942 DOI: 10.3389/fbioe.2022.921755] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022] Open
Abstract
Bioreactors are widely used in cell culture-based viral vaccine production, especially during the coronavirus disease 2019 (COVID-19) pandemic. In this context, the development and application of bioreactors can provide more efficient and cost-effective vaccine production to meet the global vaccine demand. The production of viral vaccines is inseparable from the development of upstream biological processes. In particular, exploration at the laboratory-scale is urgently required for further development. Therefore, it is necessary to evaluate the existing upstream biological processes, to enable the selection of pilot-scale conditions for academic and industrial scientists to maximize the yield and quality of vaccine development and production. Reviewing methods for optimizing the upstream process of virus vaccine production, this review discusses the bioreactor concepts, significant parameters and operational strategies related to large-scale amplification of virus. On this basis, a comprehensive analysis and evaluation of the various process optimization methods for the production of various viruses (SARS-CoV-2, Influenza virus, Tropical virus, Enterovirus, Rabies virus) in bioreactors is presented. Meanwhile, the types of viral vaccines are briefly introduced, and the established animal cell lines for vaccine production are described. In addition, it is emphasized that the co-development of bioreactor and computational biology is urgently needed to meet the challenges posed by the differences in upstream production scales between the laboratory and industry.
Collapse
Affiliation(s)
- Zhongbiao Fang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jingting Lyu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jianhua Li
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Chaonan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuxuan Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yikai Guo
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ying Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- *Correspondence: Ying Wang, ; Yanjun Zhang, ; Keda Chen,
| | - Yanjun Zhang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
- *Correspondence: Ying Wang, ; Yanjun Zhang, ; Keda Chen,
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- *Correspondence: Ying Wang, ; Yanjun Zhang, ; Keda Chen,
| |
Collapse
|
13
|
Sun Y, Huang L, Nie J, Feng K, Liu Y, Bai Z. Development of a perfusion process for serum-free adenovirus vector herpes zoster vaccine production. AMB Express 2022; 12:58. [PMID: 35567723 PMCID: PMC9107214 DOI: 10.1186/s13568-022-01398-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/04/2022] [Indexed: 11/10/2022] Open
Abstract
Herpes zoster is caused by reactivation of the varicella zoster virus (VZV). Researching and developing a herpes zoster vaccine will help to decrease the incidence of herpes zoster. To increase the bioreactor productivity, a serum-free HEK293 cell perfusion process with adenovirus vector herpes zoster (rAd-HZ) vaccine production was developed efficiently using the design of experiment (DoE) method. First, serum-free media for HEK293 cells were screened in both batch and semi-perfusion culture modes. Then, three optimal media were employed in a medium mixture design to improve cell culture performance, and the 1:1 mixture of HEK293 medium and MCD293 medium (named HM293 medium) was identified as the optimal formulation. On the basis of the HM293 medium, the relationship of critical process parameters (CPPs), including the time of infection (TOI), multiplicity of infection (MOI), pH, and critical quality attributes (CQAs) (adenovirus titer (Titer), cell-specific virus yield (CSVY), adenovirus fold expansion (Fold)) of rAd-HZ production was investigated using the DoE approach. Furthermore, the robust setpoint and design space of these CPPs were explored. Finally, the rAd-HZ production process with parameters at a robust setpoint (TOI = 7.2 × 106 cells/mL, MOI = 3.7, and pH = 7.17) was successfully scaled-up to a 3-L bioreactor with an alternating tangential flow system, yielding an adenovirus titer of 3.0 × 1010 IFU/mL, a CSVY of 4167 IFU/cells, a Fold of 1117 at 2 days post infection (dpi). The DoE approach accelerated the development of a HEK293 serum-free medium and of a robust adenovirus production process.
Collapse
|
14
|
Rosen O, Jayson A, Goldvaser M, Dor E, Monash A, Levin L, Cherry L, Lupu E, Natan N, Girshengorn M, Epstein E. Optimization of VSV-ΔG-spike production process with the Ambr15 system for a SARS-COV-2 vaccine. Biotechnol Bioeng 2022; 119:1839-1848. [PMID: 35319097 PMCID: PMC9082513 DOI: 10.1002/bit.28088] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/13/2022] [Indexed: 01/08/2023]
Abstract
To face the coronavirus disease 2019 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) virus, our institute has developed the rVSV‐ΔG‐spike vaccine, in which the glycoprotein of vesicular stomatitis virus (VSV) was replaced by the spike protein of SARS‐CoV‐2. Many process parameters can influence production yield. To maximize virus vaccine yield, each parameter should be tested independently and in combination with others. Here, we report the optimization of the production of the VSV‐ΔG‐spike vaccine in Vero cells using the Ambr15 system. This system facilitates high‐throughput screening of process parameters, as it contains 24 individually controlled, single‐use stirred‐tank minireactors. During optimization, critical parameters were tested. Those parameters included: cell densities; the multiplicity of infection; virus production temperature; medium addition and medium exchange; and supplementation of glucose in the virus production step. Virus production temperature, medium addition, and medium exchange were all found to significantly influence the yield. The optimized parameters were tested in the BioBLU 5p bioreactors production process and those that were found to contribute to the vaccine yield were integrated into the final process. The findings of this study demonstrate that an Ambr15 system is an effective tool for bioprocess optimization of vaccine production using macrocarriers and that the combination of production temperature, rate of medium addition, and medium exchange significantly improved virus yield.
Collapse
Affiliation(s)
- Osnat Rosen
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Avital Jayson
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Michael Goldvaser
- Department of Organic Chemistry, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Eyal Dor
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Arik Monash
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Lilach Levin
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Lilach Cherry
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Edith Lupu
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Niva Natan
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Meni Girshengorn
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| | - Eyal Epstein
- Department of Biotechnology, Israel Institute for Biological, Chemical and Environmental Sciences, Ness Ziona, Israel
| |
Collapse
|
15
|
Highly Efficient Purification of Recombinant VSV-∆G-Spike Vaccine against SARS-CoV-2 by Flow-Through Chromatography. BIOTECH 2021; 10:biotech10040022. [PMID: 35822796 PMCID: PMC9245476 DOI: 10.3390/biotech10040022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 01/10/2023] Open
Abstract
This study reports a highly efficient, rapid one-step purification process for the production of the recombinant vesicular stomatitis virus-based vaccine, rVSV-∆G-spike (rVSV-S), recently developed by the Israel Institute for Biological Research (IIBR) for the prevention of COVID-19. Several purification strategies are evaluated using a variety of chromatography methods, including membrane adsorbers and packed-bed ion-exchange chromatography. Cell harvest is initially treated with endonuclease, clarified, and further concentrated by ultrafiltration before chromatography purification. The use of anion-exchange chromatography in all forms results in strong binding of the virus to the media, necessitating a high salt concentration for elution. The large virus and spike protein binds very strongly to the high surface area of the membrane adsorbents, resulting in poor virus recovery (<15%), while the use of packed-bed chromatography, where the surface area is smaller, achieves better recovery (up to 33%). Finally, a highly efficient chromatography purification process with CaptoTM Core 700 resin, which does not require binding and the elution of the virus, is described. rVSV-S cannot enter the inner pores of the resin and is collected in the flow-through eluent. Purification of the rVSV-S virus with CaptoTM Core 700 resulted in viral infectivity above 85% for this step, with the efficient removal of host cell proteins, consistent with regulatory requirements. Similar results were obtained without an initial ultrafiltration step.
Collapse
|
16
|
Establishment of a Suspension MDBK Cell Line in Serum-Free Medium for Production of Bovine Alphaherpesvirus-1. Vaccines (Basel) 2021; 9:vaccines9091006. [PMID: 34579242 PMCID: PMC8473029 DOI: 10.3390/vaccines9091006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/22/2022] Open
Abstract
The Madin–Darby bovine kidney (MDBK) cell line is currently used for the production of bovine alphaherpesvirus-1 (BoHV-1) vaccine. For the purpose of vaccine manufacturing, suspension cells are preferred over adherent ones due to simplified sub-cultivation and an easier scale-up process, both of which could significantly reduce production cost. This study aimed to establish a procedure for the culture of BoHV-1 in the suspended MDBK cell line in serum-free medium. We screened several commercially available serum-free media and chose ST503 for subsequent experiments. We successfully adapted the adherent MDBK cells to suspended growth in ST503 in the absence of serum. The maximum density of suspension-adapted MDBK cells could reach 2.5 × 107 cells/mL in ST503 medium with optimal conditions. The average size of suspension-adapted cells increased to 18 ± 1 µm from 16 ± 1 µm. Moreover, we examined tumorigenicity of the suspended cells and found no sign of tumorigenicity post adaptation. Next, we developed a protocol for the culture of BoHV-1 in the cell line described above and found that ultrasonic treatment could facilitate virus release and enhance virus yield by 11-fold, with the virus titer reaching 8.0 ± 0.2 log10TCID50/mL. Most importantly, the prototype inactivated BoHV-1 vaccine we generated using the suspension cultures of MDBK cells induced neutralizing antibodies to a titer comparable to that of the commercial inactivated BoHV-1 vaccine. Overall, we established and optimized a protocol for the production of inactivated BoHV-1 vaccine in MDBK cells adapted for suspension culture, which provides insights for future large-scale manufacturing of BoHV-1 vaccine.
Collapse
|
17
|
Rosen O, Jayson A, Natan N, Monash A, Girshengorn M, Goldvaser M, Levin L, Epstein E. Novel method for quantifying cells on carriers and its demonstration during SARS-2 vaccine development. Biotechnol Bioeng 2021; 118:3811-3820. [PMID: 34110003 DOI: 10.1002/bit.27856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 11/10/2022]
Abstract
The most effective way to prevent and control infectious disease outbreak is through vaccines. The increasing use of vaccines has elevated the need to establish new manufacturing strategies. One of the major approaches is cell-based production, which creates a need for high cell density to enable higher cell production levels. This has led to development of the technology of cell carriers, including micro and macro cell carriers. To follow the production process, quantifying the number of cells on these carriers is required, as well as the tracking of their viability and proliferation. However, owing to various carriers' unique structures, tracking the cell's is challenging using current traditional assays that were originally developed for monolayers of adherent cells. The current "gold standard" method is counting cell nuclei, separating cells from the carrier, staining with crystal violet, and visually counting under a microscope. This method is tedious and counts both live and dead cells. A few other techniques were developed but were specific to the carrier type and involved specialized equipment. In this study, we describe a broadly ranging method for counting cells on carriers that was developed and employed as part of the development of severe acute respiratory syndrome coronavirus 2 vaccine. The method is based on the Alamar blue dye, a well-known, common marker for cell activity, and was found to be successful in tracking cell adsorption, cell growth, and viability on carriers. No separation of the cells from the carriers is needed, nor is any specialized equipment; the method is simple and rapid and provides comprehensive details necessary for process control of viral vaccine production in cells. This method can be easily implemented in any of a number of cell-based processes and other unique platforms for measuring the growth of encapsulated cells.
Collapse
Affiliation(s)
- Osnat Rosen
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Avital Jayson
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Niva Natan
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Arik Monash
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Meni Girshengorn
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Michael Goldvaser
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Lilach Levin
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Eyal Epstein
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona, Israel
| |
Collapse
|
18
|
Pulix M, Lukashchuk V, Smith DC, Dickson AJ. Molecular characterization of HEK293 cells as emerging versatile cell factories. Curr Opin Biotechnol 2021; 71:18-24. [PMID: 34058525 DOI: 10.1016/j.copbio.2021.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 01/27/2023]
Abstract
HEK293 cell lines are used for the production of recombinant proteins, virus-like particles and viral vectors. Recent work has generated molecular (systems level) characterisation of HEK293 variants that has enabled re-engineering of the cells towards enhanced use for manufacture-scale production of recombinant biopharmaceuticals (assessment of 'safe harbours' for gene insertion, engineering of new variants for stable, amplifiable expression). In parallel, there have been notable advances in the bioprocessing conditions (suspension adaptation, development of defined serum-free media) that offer the potential for large-scale manufacture, a feature especially important in the drive to produce viral vectors at large-scale and at commercially viable costs for gene therapy. The combination of cell-based and bioprocess-based modification of existing HEK293 cell processes, frequently informed by understandings transferred from developments with Chinese hamster ovary cell lines, seems destined to place the HEK293 cell systems firmly as a critical platform for production of future biologically based therapeutics.
Collapse
Affiliation(s)
- Michela Pulix
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, Department of Chemical Engineering & Analytical Sciences, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK; Cobra Biologics, Stephenson Building, Keele Science Park, Keele ST5 5SP, UK
| | - Vera Lukashchuk
- Cobra Biologics, Stephenson Building, Keele Science Park, Keele ST5 5SP, UK
| | - Daniel C Smith
- Cobra Biologics, Stephenson Building, Keele Science Park, Keele ST5 5SP, UK
| | - Alan J Dickson
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, Department of Chemical Engineering & Analytical Sciences, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK.
| |
Collapse
|
19
|
Kiesslich S, Kim GN, Shen CF, Kang CY, Kamen AA. Bioreactor production of rVSV-based vectors in Vero cell suspension cultures. Biotechnol Bioeng 2021; 118:2649-2659. [PMID: 33837958 PMCID: PMC8252067 DOI: 10.1002/bit.27785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/16/2021] [Accepted: 04/08/2021] [Indexed: 12/17/2022]
Abstract
The Vero cell line is the most used continuous cell line in viral vaccine manufacturing. This adherent cell culture platform requires the use of surfaces to support cell growth, typically roller bottles, or microcarriers. We have recently compared the production of rVSV‐ZEBOV on Vero cells between microcarrier and fixed‐bed bioreactors. However, suspension cultures are considered superior with regard to process scalability. Therefore, we further explore the Vero suspension system for recombinant vesicular stomatitis virus (rVSV)‐vectored vaccine production. Previously, this suspension cell line was only able to be cultivated in a proprietary medium. Here, we expand the adaptation and bioreactor cultivation to a serum‐free commercial medium. Following small‐scale optimization and screening studies, we demonstrate bioreactor productions of highly relevant vaccines and vaccine candidates against Ebola virus disease, HIV, and coronavirus disease 2019 in the Vero suspension system. rVSV‐ZEBOV, rVSV‐HIV, and rVSVInd‐msp‐SF‐Gtc can replicate to high titers in the bioreactor, reaching 3.87 × 107 TCID50/ml, 2.12 × 107 TCID50/ml, and 3.59 × 109 TCID50/ml, respectively. Furthermore, we compare cell‐specific productivities, and the quality of the produced viruses by determining the ratio of total viral particles to infectious viral particles.
Collapse
Affiliation(s)
- Sascha Kiesslich
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Gyoung N Kim
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Chun F Shen
- Human Health Therapeutics Research Center, National Research Council of Canada, Quebec, Canada
| | - C Yong Kang
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Amine A Kamen
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
20
|
Li W, Fan Z, Lin Y, Wang TY. Serum-Free Medium for Recombinant Protein Expression in Chinese Hamster Ovary Cells. Front Bioeng Biotechnol 2021; 9:646363. [PMID: 33791287 PMCID: PMC8006267 DOI: 10.3389/fbioe.2021.646363] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
At present, nearly 70% of recombinant therapeutic proteins (RTPs) are produced by Chinese hamster ovary (CHO) cells, and serum-free medium (SFM) is necessary for their culture to produce RTPs. In this review, the history and key components of SFM are first summarized, and its preparation and experimental design are described. Some small molecule compound additives can improve the yield and quality of RTP. The function and possible mechanisms of these additives are also reviewed here. Finally, the future perspectives of SFM use with CHO cells for RTP production are discussed.
Collapse
Affiliation(s)
- Weifeng Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Zhenlin Fan
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Yan Lin
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| | - Tian-Yun Wang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China.,International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
21
|
Wang L, Jiang J, Zhou T, Xue X, Cao Y. Improvement of Cerebral Ischemia-Reperfusion Injury via Regulation of Apoptosis by Exosomes Derived from BDNF-Overexpressing HEK293. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6613510. [PMID: 33763476 PMCID: PMC7952171 DOI: 10.1155/2021/6613510] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/01/2021] [Accepted: 02/25/2021] [Indexed: 12/17/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) provides neuroprotective effects towards therapeutic cerebral ischemia-reperfusion (I/R) injury. This view has been proposed by more and more evidence. However, due to the lack of permeability of the blood-brain barrier (BBB) as well as the brief half-life in serum, clinical application is not widespread. To study the participation of exosomes containing BDNF in I/R, we isolated exosomes from BDNF-overexpressing HEK293. The protective outcomes of exosomes in hypoxia/reoxygenation (H/R) experiments were determined by the use of SY-5Y cells. Exosome-BDNF therapy restrained H/R-induced apoptosis by inhibition of the reducing levels of oxidative stress and calcium ions in the cells while maintaining stable levels of mitochondrial membrane potential in brain cells damaged by I/R. We then constructed a cerebral I/R injury model using SD rats to find the function of BDNF in exosome-mediated neuroprotection. The in vivo experiments conducted established that exosomes from BDNF-overexpressing HEK293 cells improved cerebral I/R injury by concealing neuronal apoptosis. Findings gained demonstrated that BDNF is a part of preventing cerebral I/R injury due to exosome mediation by regulating the cellular internal environment and inhibiting apoptosis.
Collapse
Affiliation(s)
- Lizong Wang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Jiangsu Province, China
- Department of Neurology, The First Affiliated Hospital of Wannan Medical College, Anhui Province, China
| | - Jinghan Jiang
- Emergency Department, The First Affiliated Hospital of Wannan Medical College, Anhui Province, China
| | - Taofeng Zhou
- Department of Neurology, The First Affiliated Hospital of Wannan Medical College, Anhui Province, China
| | - Xiang Xue
- Emergency Department, The First Affiliated Hospital of Wannan Medical College, Anhui Province, China
| | - Yongjun Cao
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Jiangsu Province, China
| |
Collapse
|
22
|
Mangion M, Gélinas JF, Bakhshi Zadeh Gashti A, Azizi H, Kiesslich S, Nassoury N, Chahal PS, Kobinger G, Gilbert R, Garnier A, Gaillet B, Kamen A. Evaluation of novel HIV vaccine candidates using recombinant vesicular stomatitis virus vector produced in serum-free Vero cell cultures. Vaccine 2020; 38:7949-7955. [PMID: 33139138 DOI: 10.1016/j.vaccine.2020.10.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/09/2020] [Accepted: 10/18/2020] [Indexed: 12/20/2022]
Abstract
Acquired Immune Deficiency Syndrome (AIDS) in humans is a result of the destruction of the immune system caused by Human Immunodeficiency Virus (HIV) infection. This serious epidemic is still progressing world-wide. Despite advances in treatment, a safe and effective preventive HIV vaccine is desired to combat this disease, and to save millions of lives. However, such a vaccine is not available yet although extensive amounts of resources in research and development have been invested over three decades. In light of the recently approved Ebola virus disease vaccine based on a recombinant vesicular stomatitis virus (rVSV-ZEBOV), we present the results of our work on three novel VSV-vectored HIV vaccine candidates. We describe the design, rescue, production and purification method and evaluate their immunogenicity in mice prior to preclinical studies that will be performed in non-human primates. The production of each of the three candidate vaccines (rVSV-B6-NL4.3Env/SIVtm, rVSV-B6-NL4.3Env/Ebtm and rVSV-B6-A74Env(PN6)/SIVtm) was evaluated in small scale in Vero cells and it was found that production kinetics on Vero cells vary depending on the HIV gp surface protein used. Purified virus preparations complied with the WHO restrictions for the residual DNA and host cell protein contents. Finally, when administered to mice, all three rVSV-HIV vaccine candidates induced an HIV gp140-specific antibody response.
Collapse
Affiliation(s)
- Mathias Mangion
- Département de génie chimique, Université Laval, Québec, QC, Canada
| | | | | | - Hiva Azizi
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Quebec, QC, Canada
| | - Sascha Kiesslich
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Nasha Nassoury
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Parminder S Chahal
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Gary Kobinger
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Quebec, QC, Canada
| | - Rénald Gilbert
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Alain Garnier
- Département de génie chimique, Université Laval, Québec, QC, Canada
| | - Bruno Gaillet
- Département de génie chimique, Université Laval, Québec, QC, Canada
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, QC, Canada.
| |
Collapse
|
23
|
Gélinas JF, Kiesslich S, Gilbert R, Kamen AA. Titration methods for rVSV-based vaccine manufacturing. MethodsX 2020; 7:100806. [PMID: 32195130 PMCID: PMC7078374 DOI: 10.1016/j.mex.2020.100806] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/22/2020] [Indexed: 11/21/2022] Open
Abstract
The recombinant Vesicular Stomatitis Virus (rVSV) is an emerging platform for viral vector-based vaccines. Promising results have been reported in clinical trials for the rVSV-ZEBOV vaccine for Ebola virus disease prevention. In this study, we describe the titration tools elaborated to assess the titre of rVSV-ZEBOV productions. • A streamlined Median Tissue Culture Infectious Dose (TCID50) assay to determine the infectious titer of this vaccine was established. • A digital polymerase chain reaction (dPCR) assay to assess the total number of viral particles present in cell-free culture supernatants of rVSV productions was developed. • These assays are used to titre rVSV-ZEBOV samples and characterize the ratio of total particles to infectious units for monitoring process robustness and product quality attributes and can be used to titre samples generated in the production of further rVSV vectors.
Collapse
Affiliation(s)
- Jean-François Gélinas
- Department of Bioengineering, McGill University, 3480 University, Montréal, Québec H3A 0E9, Canada
| | - Sascha Kiesslich
- Department of Bioengineering, McGill University, 3480 University, Montréal, Québec H3A 0E9, Canada
| | - Rénald Gilbert
- Department of Bioengineering, McGill University, 3480 University, Montréal, Québec H3A 0E9, Canada
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Amine A. Kamen
- Department of Bioengineering, McGill University, 3480 University, Montréal, Québec H3A 0E9, Canada
- Corresponding author.
| |
Collapse
|
24
|
Kiesslich S, Vila-Chã Losa JP, Gélinas JF, Kamen AA. Serum-free production of rVSV-ZEBOV in Vero cells: Microcarrier bioreactor versus scale-X™ hydro fixed-bed. J Biotechnol 2020; 310:32-39. [DOI: 10.1016/j.jbiotec.2020.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/12/2019] [Accepted: 01/28/2020] [Indexed: 12/29/2022]
|