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] [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
|
Pelz L, Dogra T, Marichal-Gallardo P, Hein MD, Hemissi G, Kupke SY, Genzel Y, Reichl U. Production of antiviral "OP7 chimera" defective interfering particles free of infectious virus. Appl Microbiol Biotechnol 2024; 108:97. [PMID: 38229300 DOI: 10.1007/s00253-023-12959-6] [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: 08/21/2023] [Revised: 10/26/2023] [Accepted: 11/05/2023] [Indexed: 01/18/2024]
Abstract
Defective interfering particles (DIPs) of influenza A virus (IAV) are suggested for use as broad-spectrum antivirals. We discovered a new type of IAV DIP named "OP7" that carries point mutations in its genome segment (Seg) 7 instead of a deletion as in conventional DIPs (cDIPs). Recently, using genetic engineering tools, we generated "OP7 chimera DIPs" that carry point mutations in Seg 7 plus a deletion in Seg 1. Together with cDIPs, OP7 chimera DIPs were produced in shake flasks in the absence of infectious standard virus (STV), rendering UV inactivation unnecessary. However, only part of the virions harvested were OP7 chimera DIPs (78.7%) and total virus titers were relatively low. Here, we describe the establishment of an OP7 chimera DIP production process applicable for large-scale production. To increase total virus titers, we reduced temperature from 37 to 32 °C during virus replication. Production of almost pure OP7 chimera DIP preparations (99.7%) was achieved with a high titer of 3.24 log10(HAU/100 µL). This corresponded to an 11-fold increase relative to the initial process. Next, this process was transferred to a stirred tank bioreactor resulting in comparable yields. Moreover, DIP harvests purified and concentrated by steric exclusion chromatography displayed an increased interfering efficacy in vitro. Finally, a perfusion process with perfusion rate control was established, resulting in a 79-fold increase in total virus yields compared to the original batch process in shake flasks. Again, a very high purity of OP7 chimera DIPs was obtained. This process could thus be an excellent starting point for good manufacturing practice production of DIPs for use as antivirals. KEY POINTS: • Scalable cell culture-based process for highly effective antiviral OP7 chimera DIPs • Production of almost pure OP7 chimera DIPs in the absence of infectious virus • Perfusion mode production and purification train results in very high titers.
Collapse
Affiliation(s)
- Lars Pelz
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Tanya Dogra
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Pavel Marichal-Gallardo
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Marc Dominique Hein
- Otto Von Guericke University Magdeburg, Bioprocess Engineering, Magdeburg, Germany
| | - Ghada Hemissi
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Sascha Young Kupke
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany.
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany.
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
- Otto Von Guericke University Magdeburg, Bioprocess Engineering, Magdeburg, Germany
| |
Collapse
|
3
|
Park A, Lee YJ, Jo E, Park GH, Heo SY, Koh EJ, Lee SH, Cha SH, Heo SJ. Serum-Free Media Formulation Using Marine Microalgae Extracts and Growth Factor Cocktails for Madin-Darby Canine Kidney and Vero Cell Cultures. Int J Mol Sci 2024; 25:9881. [PMID: 39337368 PMCID: PMC11432547 DOI: 10.3390/ijms25189881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The development of serum-free media (SFM) is critical to advance cell culture techniques used in viral vaccine production and address the ethical concerns and contamination risks associated with fetal bovine serum (FBS). This study evaluated the effects of marine microalgal extracts and growth factor cocktails on the activity of Madin-Darby canine kidney (MDCK) and Vero cells. Five marine microalgal species were used: Spirulina platensis (SP), Dunaliella salina (DS), Haematococcus pluvialis (HP), Nannochloropsis salina (NS), and Tetraselmis sp. (TS). DS and SP extracts significantly increased the proliferation rate of both MDCK and Vero cells. DS had a proliferation rate of 149.56% and 195.50% in MDCK and Vero cells, respectively, compared with that in serum-free medium (SFM). Notably, DS and SP extracts significantly increased superoxide dismutase (SOD) activity, which was 118.61% in MDCK cells and 130.08% in Vero cells for DS, and 108.72% in MDCK cells and 125.63% in Vero cells for SP, indicating a reduction in intracellular oxidative stress. Marine microalgal extracts, especially DS and SP, are feasible alternatives to FBS in cell culture as they promote cell proliferation, ensure safety, and supply essential nutrients while reducing oxidative stress.
Collapse
Affiliation(s)
- Areumi Park
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
| | - Yeon-Ji Lee
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
| | - Eunyoung Jo
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
| | - Gun-Hoo Park
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
| | - Seong-Yeong Heo
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
- Department of Marine Technology & Convergence Engineering (Marine Biotechnology), University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Eun-Jeong Koh
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
| | - Seung-Hong Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Seon-Heui Cha
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 32158, Republic of Korea
| | - Soo-Jin Heo
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
- Department of Marine Technology & Convergence Engineering (Marine Biotechnology), University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
4
|
Rüdiger D, Piasecka J, Küchler J, Pontes C, Laske T, Kupke SY, Reichl U. Mathematical model calibrated to in vitro data predicts mechanisms of antiviral action of the influenza defective interfering particle "OP7". iScience 2024; 27:109421. [PMID: 38523782 PMCID: PMC10959662 DOI: 10.1016/j.isci.2024.109421] [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: 08/23/2023] [Revised: 02/08/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Defective interfering particles (DIPs) are regarded as potent broad-spectrum antivirals. We developed a mathematical model that describes intracellular co-infection dynamics of influenza standard virus (STV) and "OP7", a new type of influenza DIP discovered recently. Based on experimental data from in vitro studies to calibrate the model and confirm its predictions, we deduce OP7's mechanisms of interference, which were yet unknown. Simulations suggest that the "superpromoter" on OP7 genomic viral RNA enhances its replication and results in a depletion of viral proteins. This reduces STV genomic RNA replication, which appears to constitute an antiviral effect. Further, a defective viral protein (M1-OP7) likely causes the deficiency of OP7's replication. It appears unable to bind to genomic viral RNAs to facilitate their nuclear export, a critical step in the viral life cycle. An improved understanding of OP7's antiviral mechanism is crucial toward application in humans as a prospective antiviral treatment strategy.
Collapse
Affiliation(s)
- Daniel Rüdiger
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Saxony-Anhalt, Germany
| | - Julita Piasecka
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Saxony-Anhalt, Germany
| | - Jan Küchler
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Saxony-Anhalt, Germany
| | - Carolina Pontes
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Saxony-Anhalt, Germany
| | - Tanja Laske
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Saxony-Anhalt, Germany
- Institute for Computational Systems Biology, University of Hamburg, 20148 Hamburg, Germany
| | - Sascha Y. Kupke
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Saxony-Anhalt, Germany
| | - Udo Reichl
- Department of Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, 39106 Magdeburg, Saxony-Anhalt, Germany
- Chair of Bioprocess Engineering, Otto-von-Guericke University, 39106 Magdeburg, Saxony-Anhalt, Germany
| |
Collapse
|
5
|
Zabrodskaya Y, Tsvetkov V, Shurygina AP, Vasyliev K, Shaldzhyan A, Gorshkov A, Kuklin A, Fedorova N, Egorov V. How the immune mousetrap works: Structural evidence for the immunomodulatory action of a peptide from influenza NS1 protein. Biophys Chem 2024; 307:107176. [PMID: 38219420 DOI: 10.1016/j.bpc.2024.107176] [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: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 01/16/2024]
Abstract
One of the critical stages of the T-cell immune response is the dimerization of the intramembrane domains of T-cell receptors (TCR). Structural similarities between the immunosuppressive domains of viral proteins and the transmembrane domains of TCR have led several authors to hypothesize the mechanism of immune response suppression by highly pathogenic viruses: viral proteins embed themselves in the membrane and act on the intramembrane domain of the TCRalpha subunit, hindering its functional oligomerization. It has also been suggested that this mechanism is used by influenza A virus in NS1-mediated immunosuppression. We have shown that the peptide corresponding to the primary structure of the potential immunosuppressive domain of NS1 protein (G51) can reduce concanavalin A-induced proliferation of PBMC cells, as well as in vitro, G51 can affect the oligomerization of the core peptide corresponding to the intramembrane domain of TCR, using AFM and small-angle neutron scattering. The results obtained using in cellulo and in vitro model systems suggest the presence of functional interaction between the NS1 fragment and the intramembrane domain of the TCR alpha subunit. We have proposed a possible scheme for such interaction obtained by computer modeling. This suggests the existence of another NS1-mediated mechanism of immunosuppression in influenza.
Collapse
Affiliation(s)
- Yana Zabrodskaya
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, St. Petersburg 194064, Russia; Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia.
| | - Vladimir Tsvetkov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia; Federal Research and Clinical Center for Physical Chemical Medicine, 1a Ulitsa Malaya Pirogovskaya, Moscow 119435, Russia; Center for Mathematical Modeling in Drug Development, I.M. Sechenov First Moscow State Medical University, Moscow 119146, Russia
| | - Anna-Polina Shurygina
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia
| | - Kirill Vasyliev
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia
| | - Aram Shaldzhyan
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia
| | - Andrey Gorshkov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 15/17 Ulitsa Prof. Popova, St. Petersburg 197376, Russia
| | - Alexander Kuklin
- International Intergovernmental Organization Joint Institute for Nuclear Research, 6 Ulitsa Joliot-Curie, Dubna 141980, Russia; Moscow Institute of Physics and Technology (State University), 9 Institutskiy pereulok, 141701 Dolgoprudny, Moscow Region, Russia
| | - Natalya Fedorova
- Petersburg Nuclear Physics Institute Named by B. P. Konstantinov of National Research Center, Kurchatov Institute, 1 mkr. Orlova Roshcha, Gatchina 188300, Russia
| | - Vladimir Egorov
- Institute of Experimental Medicine, 12 Ulitsa Akademika Pavlova, St. Petersburg 197376, Russia
| |
Collapse
|
6
|
Zinnecker T, Badri N, Araujo D, Thiele K, Reichl U, Genzel Y. From single-cell cloning to high-yield influenza virus production - implementing advanced technologies in vaccine process development. Eng Life Sci 2024; 24:2300245. [PMID: 38584687 PMCID: PMC10991716 DOI: 10.1002/elsc.202300245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 04/09/2024] Open
Abstract
Innovations in viral vaccine manufacturing are crucial for pandemic preparedness and to meet ever-rising global demands. For influenza, however, production still mainly relies on technologies established decades ago. Although modern production shifts from egg-based towards cell culture technologies, the full potential has not yet been fully exploited. Here, we evaluate whether implementation of state-of-the-art technologies for cell culture-based recombinant protein production are capable to challenge outdated approaches in viral vaccine process development. For this, a fully automated single-cell cloning strategy was established to generate monoclonal suspension Madin-Darby canine kidney (MDCK) cells. Among selected cell clones, we could observe distinct metabolic and growth characteristics, with C59 reaching a maximum viable cell concentration of 17.3 × 106 cells/mL and low doubling times in batch mode. Screening for virus production using a panel of human vaccine-relevant influenza A and B viruses in an ambr15 system revealed high titers with yields competing or even outperforming available MDCK cell lines. With C113, we achieved cell-specific virus yields of up to 25,000 virions/cell, making this cell clone highly attractive for vaccine production. Finally, we confirmed process performance at a 50-fold higher working volume. In summary, we present a scalable and powerful approach for accelerated development of high-yield influenza virus production in chemically defined medium starting from a single cell.
Collapse
Affiliation(s)
- Tilia Zinnecker
- Max Planck Institute for Dynamics of Complex Technical SystemsMagdeburgGermany
| | | | - Diogo Araujo
- Sartorius Stedim Biotech S.A.Aubagne CedexFrance
| | | | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical SystemsMagdeburgGermany
- Bioprocess EngineeringOtto‐von‐Guericke UniversityMagdeburgGermany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical SystemsMagdeburgGermany
| |
Collapse
|
7
|
Kim JY, Park SY, Lee G, Park SH, Jin JS, Kim D, Park JH, Jeong SY, Ko YJ. Determination of Optimal Antigen Yield and Virus Inactivation Conditions for the Production of the Candidate Foot-and-Mouth Disease Recombinant Vaccine Strain Asia1 Shamir-R in a Bioreactor. Viruses 2024; 16:457. [PMID: 38543822 PMCID: PMC10974838 DOI: 10.3390/v16030457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 05/23/2024] Open
Abstract
Since the foot-and-mouth disease (FMD) outbreak in South Korea in 2010-2011, vaccination policies utilizing inactivated FMD vaccines composed of types O and A have been implemented nationwide. However, because type Asia1 occurred in North Korea in 2007 and intermittently in neighboring countries, the risk of type Asia1 introduction cannot be ruled out. This study evaluated the antigen yield and viral inactivation kinetics of the recombinant Asia1 Shamir vaccine strain (Asia1 Shamir-R). When Asia1 Shamir-R was proliferated in shaking flasks (1 L), a 2 L bioreactor (1 L), and a wave bioreactor (25 L), the antigen yields were 7.5 μg/mL, 5.2 μg/mL, and 3.8 μg/mL, respectively. The optimal FMDV inactivation conditions were 2 mM BEI at 26 °C and 1.0 mM BEI at 37 °C. There was no antigen loss due to BEI treatment, and only a decrease in antigen levels was observed during storage. The sera from pigs immunized with antigen derived from a bioreactor exhibited a neutralizing antibody titer of approximately 1/1000 against Asia1 Shamir and Asia1/MOG/05 viruses; therefore, Asia1 Shamir-R is expected to provide sufficient protection against both viruses. If an FMD vaccine production facility is established, this Asia1 Shamir-R can be employed for domestic antigen banks in South Korea.
Collapse
Affiliation(s)
- Jae Young Kim
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
- Department of Biomedical Science, Graduate School, Catholic University of Daegu, Daegu 38430, Republic of Korea;
| | - Sun Young Park
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
| | - Gyeongmin Lee
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
| | - Sang Hyun Park
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
| | - Jong-Sook Jin
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
| | - Dohyun Kim
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
| | - Jong-Hyeon Park
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
| | - Seong-Yun Jeong
- Department of Biomedical Science, Graduate School, Catholic University of Daegu, Daegu 38430, Republic of Korea;
| | - Young-Joon Ko
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Republic of Korea; (J.Y.K.); (G.L.); (D.K.)
| |
Collapse
|
8
|
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] [Track Full Text] [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
|
9
|
Kim JY, Park SY, Park SH, Lee G, Jin JS, Kim D, Park JH, Jeong SY, Ko YJ. Evaluation of Foot-and-Mouth Disease (FMD) Virus Asia1 Genotype-V as an FMD Vaccine Candidate: Study on Vaccine Antigen Production Yield and Inactivation Kinetics. Vaccines (Basel) 2024; 12:185. [PMID: 38400168 PMCID: PMC10892639 DOI: 10.3390/vaccines12020185] [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: 01/15/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
South Korea has experienced outbreaks of foot-and-mouth disease (FMD) of serotypes O and A, leading to nationwide vaccination with a bivalent vaccine. Since the FMD virus (FMDV) Asia1 group-V genotype occurred in North Korea in 2007, an Asia1/MOG/05 vaccine strain belonging to the Asia1 group-V genotype was developed using a genetic recombination method (Asia1/MOG/05-R). This study aimed to evaluate the antigen productivity and viral inactivation kinetics of Asia1/MOG/05-R to assess its commercial viability. The antigen yield of Asia1/MOG/05-R produced in flasks and bioreactors was approximately 4.0 μg/mL. Binary ethylenimine (BEI) inactivation kinetics of Asia1/MOG/05-R showed that 2 mM and 1.0 mM BEI treatment at 26 °C and 37 °C, respectively, resulted in a virus titer <10-7 TCID50/mL within 24 h, meeting the inactivation kinetics criteria. During incubation at 26 °C and 37 °C, 10% antigen loss occurred, but not due to BEI treatment. When pigs were inoculated twice with the Asia1/MOG/05-R antigen, the virus neutralization titer increased to approximately 1:1000; therefore, it can sufficiently protect against Asia1/MOG/05-R and Asia1 Shamir viruses. The Asia1/MOG/05-R will be useful as a vaccine strain for domestic antigen banks.
Collapse
Affiliation(s)
- Jae Young Kim
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
- Department of Biomedical Science, Graduate School, Catholic University of Daegu, Daegu 38430, Republic of Korea;
| | - Sun Young Park
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
| | - Sang Hyun Park
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
| | - Gyeongmin Lee
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
| | - Jong-Sook Jin
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
| | - Dohyun Kim
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
| | - Jong-Hyeon Park
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
| | - Seong-Yun Jeong
- Department of Biomedical Science, Graduate School, Catholic University of Daegu, Daegu 38430, Republic of Korea;
| | - Young-Joon Ko
- Center for FMD Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si 177, Republic of Korea; (J.Y.K.); (S.Y.P.); (S.H.P.); (G.L.); (J.-S.J.); (D.K.); (J.-H.P.)
| |
Collapse
|
10
|
Lomont JP, Smith JP. In situ process analytical technology for real time viable cell density and cell viability during live-virus vaccine production. Int J Pharm 2024; 649:123630. [PMID: 38040394 DOI: 10.1016/j.ijpharm.2023.123630] [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: 09/15/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 12/03/2023]
Abstract
Viable cell density (VCD) and cell viability (CV) are key performance indicators of cell culture processes in biopharmaceutical production of biologics and vaccines. Traditional methods for monitoring VCD and CV involve offline cell counting assays that are both labor intensive and prone to high variability, resulting in sparse sampling and uncertainty in the obtained data. Process analytical technology (PAT) approaches offer a means to address these challenges. Specifically, in situ probe-based measurements of dielectric spectroscopy (also commonly known as capacitance) can characterize VCD and CV continuously in real time throughout an entire process, enabling robust process characterization. In this work, we propose in situ dielectric spectroscopy as a PAT tool for real time analysis of live-virus vaccine (LVV) production. Dielectric spectroscopy was collected across 25 discreet frequencies, offering a thorough evaluation of the proposed technology. Correlation of this PAT methodology to traditional offline cell counting assays was performed, in which VCD and CV were both successfully predicted using dielectric spectroscopy. Both univariate and multivariate data analysis approaches were evaluated for their potential to establish correlation between the in situ dielectric spectroscopy and offline measurements. Univariate analysis strategies are presented for optimal single frequency selection. Multivariate analysis, in the form of partial least squares (PLS) regression, produced significantly higher correlations between dielectric spectroscopy and offline VCD and CV data, as compared to univariate analysis. Specifically, by leveraging multivariate analysis of dielectric information from all 25 spectroscopic frequencies measured, PLS models performed significantly better than univariate models. This is particularly evident during cell death, where tracking VCD and CV have historically presented the greatest challenge. The results of this work demonstrate the potential of both single and multiple frequency dielectric spectroscopy measurements for enabling robust LVV process characterization, suggesting that broader application of in situ dielectric spectroscopy as a PAT tool in LVV processes can provide significantly improved process understanding. To the best of our knowledge, this is the first report of in situ dielectric spectroscopy with multivariate analysis to successfully predict VCD and CV in real time during live virus-based vaccine production.
Collapse
Affiliation(s)
- Justin P Lomont
- Analytical Research & Development, MRL, Merck & Co., Inc., West Point, PA 19486, USA.
| | - Joseph P Smith
- Process Research & Development, MRL, Merck & Co., Inc., West Point, PA 19486, USA.
| |
Collapse
|
11
|
Eshchenko N, Sergeeva M, Zhuravlev E, Kudria K, Goncharova E, Komissarov A, Stepanov G. A Knockout of the IFITM3 Gene Increases the Sensitivity of WI-38 VA13 Cells to the Influenza A Virus. Int J Mol Sci 2024; 25:625. [PMID: 38203797 PMCID: PMC10778886 DOI: 10.3390/ijms25010625] [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: 12/07/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
One of the ways to regulate the sensitivity of human cells to the influenza virus is to knock out genes of the innate immune response. Promising targets for the knockout are genes of the interferon-inducible transmembrane protein (IFITM) family, in particular the IFITM3 gene, whose product limits the entry of a virus into the cell by blocking the fusion of the viral and endosomal membranes. In this study, by means of genome-editing system CRISPR/Cas9, monoclonal cell lines with an IFITM3 knockout were obtained based on WI-38 VA13 cells (human origin). It was found that such cell lines are more sensitive to infection by influenza A viruses of various subtypes. Nevertheless, this feature is not accompanied by an increased titer of newly formed viral particles in a culture medium.
Collapse
Affiliation(s)
- Natalya Eshchenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| | - Mariia Sergeeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg 197376, Russia;
| | - Evgenii Zhuravlev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| | - Kira Kudria
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg 197376, Russia;
| | - Elena Goncharova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| | - Andrey Komissarov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg 197376, Russia;
| | - Grigory Stepanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (N.E.); (M.S.); (E.G.); (A.K.); (G.S.)
| |
Collapse
|
12
|
Dogra T, Pelz L, Boehme JD, Kuechler J, Kershaw O, Marichal-Gallardo P, Baelkner M, Hein MD, Gruber AD, Benndorf D, Genzel Y, Bruder D, Kupke SY, Reichl U. Generation of "OP7 chimera" defective interfering influenza A particle preparations free of infectious virus that show antiviral efficacy in mice. Sci Rep 2023; 13:20936. [PMID: 38017026 PMCID: PMC10684881 DOI: 10.1038/s41598-023-47547-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023] Open
Abstract
Influenza A virus (IAV) defective interfering particles (DIPs) are considered as new promising antiviral agents. Conventional DIPs (cDIPs) contain a deletion in the genome and can only replicate upon co-infection with infectious standard virus (STV), during which they suppress STV replication. We previously discovered a new type of IAV DIP "OP7" that entails genomic point mutations and displays higher antiviral efficacy than cDIPs. To avoid safety concerns for the medical use of OP7 preparations, we developed a production system that does not depend on infectious IAV. We reconstituted a mixture of DIPs consisting of cDIPs and OP7 chimera DIPs, in which both harbor a deletion in their genome. To complement the defect, the deleted viral protein is expressed by the suspension cell line used for production in shake flasks. Here, DIP preparations harvested are not contaminated with infectious virions, and the fraction of OP7 chimera DIPs depended on the multiplicity of infection. Intranasal administration of OP7 chimera DIP material was well tolerated in mice. A rescue from an otherwise lethal IAV infection and no signs of disease upon OP7 chimera DIP co-infection demonstrated the remarkable antiviral efficacy. The clinical development of this new class of broad-spectrum antiviral may contribute to pandemic preparedness.
Collapse
Affiliation(s)
- Tanya Dogra
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Lars Pelz
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Julia D Boehme
- Institute of Medical Microbiology, Infection Prevention and Control, Infection Immunology Group, Health Campus Immunology, Infectiology and Inflammation, Otto Von Guericke University Magdeburg, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jan Kuechler
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Pavel Marichal-Gallardo
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Maike Baelkner
- Institute of Medical Microbiology, Infection Prevention and Control, Infection Immunology Group, Health Campus Immunology, Infectiology and Inflammation, Otto Von Guericke University Magdeburg, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marc D Hein
- Bioprocess Engineering, Otto Von Guericke University Magdeburg, Magdeburg, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Dirk Benndorf
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
- Bioprocess Engineering, Otto Von Guericke University Magdeburg, Magdeburg, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Dunja Bruder
- Institute of Medical Microbiology, Infection Prevention and Control, Infection Immunology Group, Health Campus Immunology, Infectiology and Inflammation, Otto Von Guericke University Magdeburg, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sascha Y Kupke
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany.
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
- Bioprocess Engineering, Otto Von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
13
|
Malenovská H. Ruxolitinib accelerates influenza A virus adaptation in the Madin-Darby canine kidney (MDCK) cell line. J Appl Microbiol 2023; 134:lxad232. [PMID: 37816667 DOI: 10.1093/jambio/lxad232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023]
Abstract
AIM To investigate the effect of ruxolitinib medium supplement, separately and in combination with trypsin, on influenza A virus (IAV) adaptation and propagation in the Madin-Darby canine kidney (MDCK) cell line. METHODS AND RESULTS Two consecutive passages of three egg-based IAV strains were performed in the MDCK cell line with medium (a) without additives; (b) with a combination of ruxolitinib and trypsin; (c) with ruxolitinib; and (d) trypsin. Adaptation without a medium additive failed in both passages. After a single passage, the probability of the IAV adaptation was highly significantly influenced by the type of additive (binomial generalized linear model, χ22 = 23.84, P < 0.00001). The highest probability of adaptation was achieved with the combination of ruxolitinib and trypsin, followed by ruxolitinib alone and trypsin. After the two consecutive passages, the influence of the type of medium additive on the probability of virus adaptation was no longer significant. In two of three IAV MDCK-adapted strains, the type of medium additive had no significant influence on virus yields. CONCLUSION Ruxolitinib accelerates the adaptation of IAV in the MDCK cell line both individually and together with trypsin.
Collapse
Affiliation(s)
- Hana Malenovská
- Collection of Animal Pathogenic Microorganisms, Veterinary Research Institute,Hudcova 296/70, 621 00 Brno-Medlánky, Czech Republic
| |
Collapse
|
14
|
Hein MD, Kazenmaier D, van Heuvel Y, Dogra T, Cattaneo M, Kupke SY, Stitz J, Genzel Y, Reichl U. Production of retroviral vectors in continuous high cell density culture. Appl Microbiol Biotechnol 2023; 107:5947-5961. [PMID: 37542575 PMCID: PMC10485120 DOI: 10.1007/s00253-023-12689-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 08/07/2023]
Abstract
Retroviral vectors derived from murine leukemia virus (MLV) are used in somatic gene therapy applications e.g. for genetic modification of hematopoietic stem cells. Recently, we reported on the establishment of a suspension viral packaging cell line (VPC) for the production of MLV vectors. Human embryonic kidney 293-F (HEK293-F) cells were genetically modified for this purpose using transposon vector technology. Here, we demonstrate the establishment of a continuous high cell density (HCD) process using this cell line. First, we compared different media regarding the maximum achievable viable cell concentration (VCC) in small scale. Next, we transferred this process to a stirred tank bioreactor before we applied intensification strategies. Specifically, we established a perfusion process using an alternating tangential flow filtration system. Here, VCCs up to 27.4E + 06 cells/mL and MLV vector titers up to 8.6E + 06 transducing units/mL were achieved. Finally, we established a continuous HCD process using a tubular membrane for cell retention and continuous viral vector harvesting. Here, the space-time yield was 18-fold higher compared to the respective batch cultivations. Overall, our results clearly demonstrate the feasibility of HCD cultivations for high yield production of viral vectors, especially when combined with continuous viral vector harvesting. KEY POINTS: • A continuous high cell density process for MLV vector production was established • The tubular cell retention membrane allowed for continuous vector harvesting • The established process had a 18-fold higher space time yield compared to a batch.
Collapse
Affiliation(s)
- Marc D Hein
- Chair of Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Daniel Kazenmaier
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Faculty of Biotechnology, University of Applied Sciences Mannheim, Mannheim, Germany
| | - Yasemin van Heuvel
- Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Leverkusen, Germany
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Tanya Dogra
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | | | - Sascha Y Kupke
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Jörn Stitz
- Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Leverkusen, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.
| | - Udo Reichl
- Chair of Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| |
Collapse
|
15
|
van Heuvel Y, Schatz S, Hein M, Dogra T, Kazenmaier D, Tschorn N, Genzel Y, Stitz J. Novel suspension retroviral packaging cells generated by transposition using transposase encoding mRNA advance vector yields and enable production in bioreactors. Front Bioeng Biotechnol 2023; 11:1076524. [PMID: 37082212 PMCID: PMC10112512 DOI: 10.3389/fbioe.2023.1076524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
To date, the establishment of high-titer stable viral packaging cells (VPCs) at large scale for gene therapeutic applications is very time- and cost-intensive. Here we report the establishment of three human suspension 293-F-derived ecotropic MLV-based VPCs. The classic stable transfection of an EGFP-expressing transfer vector resulted in a polyclonal VPC pool that facilitated cultivation in shake flasks of 100 mL volumes and yielded high functional titers of more than 1 × 106 transducing units/mL (TU/mL). When the transfer vector was flanked by transposon terminal inverted repeats (TIRs) and upon co-transfection of a plasmid encoding for the transposase, productivities could be slightly elevated to more than 3 × 106 TU/mL. In contrast and using mRNA encoding for the transposase, as a proof of concept, productivities were drastically improved by more than ten-fold exceeding 5 × 107 TU/mL. In addition, these VPC pools were generated within only 3 weeks. The production volume was successfully scaled up to 500 mL employing a stirred-tank bioreactor (STR). We anticipate that the stable transposition of transfer vectors employing transposase transcripts will be of utility for the future establishment of high-yield VPCs producing pseudotype vector particles with a broader host tropism on a large scale.
Collapse
Affiliation(s)
- Yasemin van Heuvel
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Stefanie Schatz
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Marc Hein
- Chair of Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Tanya Dogra
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Daniel Kazenmaier
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
- Faculty of Biotechnology, University of Applied Sciences Mannheim, Mannheim, Germany
| | - Natalie Tschorn
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Jörn Stitz
- Research Group Medical Biotechnology and Bioengineering, Faculty of Applied Natural Sciences, University of Applied Sciences Cologne, Campus Leverkusen, Cologne, Germany
- *Correspondence: Jörn Stitz,
| |
Collapse
|
16
|
Effect of Dexamethasone on the Expression of the α2,3 and α2,6 Sialic Acids in Epithelial Cell Lines. Pathogens 2022; 11:pathogens11121518. [PMID: 36558852 PMCID: PMC9788320 DOI: 10.3390/pathogens11121518] [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: 10/19/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
N-acetylneuraminic acid linked to galactose by α2,6 and α2,3 linkages (Siaα2,6 and Siaα2,3) is expressed on glycoconjugates of animal tissues, where it performs multiple biological functions. In addition, these types of sialic acid residues are the main targets for the binding and entry of influenza viruses. Here we used fluorochrome-conjugated Sambuccus nigra, Maackia amurensis, and peanut lectins for the simultaneous detection of Siaα2,3 and Siaα2,6 and galactosyl residues by two-color flow cytometry on A549 cells, a human pneumocyte cell line used for in vitro studies of the infection by influenza viruses, as well as on Vero and MDCK cell lines. The dexamethasone (DEX) glucocorticoid (GC), a widely used anti-inflammatory compound, completely abrogated the expression of Siaα2,3 in A549 cells and decreased its expression in Vero and MDCK cells; in contrast, the expression of Siaα2,6 was increased in the three cell lines. These observations indicate that DEX can be used for the study of the mechanism of sialylation of cell membrane molecules. Importantly, DEX may change the tropism of avian and human/pig influenza viruses and other infectious agents to animal and human epithelial cells.
Collapse
|
17
|
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: 6] [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
|
18
|
Göbel S, Kortum F, Chavez KJ, Jordan I, Sandig V, Reichl U, Altomonte J, Genzel Y. Cell-line screening and process development for a fusogenic oncolytic virus in small-scale suspension cultures. Appl Microbiol Biotechnol 2022; 106:4945-4961. [PMID: 35767011 PMCID: PMC9329169 DOI: 10.1007/s00253-022-12027-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/09/2022] [Accepted: 06/10/2022] [Indexed: 11/27/2022]
Abstract
Abstract
Oncolytic viruses (OVs) represent a novel class of immunotherapeutics under development for the treatment of cancers. OVs that express a cognate or transgenic fusion protein is particularly promising as their enhanced intratumoral spread via syncytia formation can be a potent mechanism for tumor lysis and induction of antitumor immune responses. Rapid and efficient fusion of infected cells results in cell death before high titers are reached. Although this is an attractive safety feature, it also presents unique challenges for large-scale clinical-grade manufacture of OVs. Here we evaluate the use of four different suspension cell lines for the production of a novel fusogenic hybrid of vesicular stomatitis virus and Newcastle disease virus (rVSV-NDV). The candidate cell lines were screened for growth, metabolism, and virus productivity. Permissivity was evaluated based on extracellular infectious virus titers and cell-specific virus yields (CSVYs). For additional process optimizations, virus adaptation and multiplicity of infection (MOI) screenings were performed and confirmed in a 1 L bioreactor. BHK-21 and HEK293SF cells infected at concentrations of 2 × 106 cells/mL were identified as promising candidates for rVSV-NDV production, leading to infectious titers of 3.0 × 108 TCID50/mL and 7.5 × 107 TCID50/mL, and CSVYs of 153 and 9, respectively. Compared to the AGE1.CR.pIX reference produced in adherent cultures, oncolytic potency was not affected by production in suspension cultures and possibly even increased in cultures of HEK293SF and AGE1.CR.pIX. Our study describes promising suspension cell-based processes for efficient large-scale manufacturing of rVSV-NDV. Key points • Cell contact-dependent oncolytic virus (OV) replicates in suspension cells. • Oncolytic potency is not encompassed during suspension cultivation. • Media composition, cell line, and MOI are critical process parameters for OV production. • The designed process is scalable and shows great promise for manufacturing clinical-grade material. Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-12027-5.
Collapse
Affiliation(s)
- Sven Göbel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Fabian Kortum
- Department of Internal Medicine II, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Karim Jaén Chavez
- Department of Internal Medicine II, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Ingo Jordan
- ProBioGen AG, Herbert-Bayer-Str. 8, 13086, Berlin, Germany
| | - Volker Sandig
- ProBioGen AG, Herbert-Bayer-Str. 8, 13086, Berlin, Germany
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106, Magdeburg, Germany
- Chair for Bioprocess Engineering, Otto-Von-Guericke-University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Jennifer Altomonte
- Department of Internal Medicine II, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106, Magdeburg, Germany.
| |
Collapse
|
19
|
Expression of the Heterotrimeric GP2/GP3/GP4 Spike of an Arterivirus in Mammalian Cells. Viruses 2022; 14:v14040749. [PMID: 35458479 PMCID: PMC9030998 DOI: 10.3390/v14040749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/05/2023] Open
Abstract
Equine arteritis virus (EAV), an enveloped positive-strand RNA virus, is an important pathogen of horses and the prototype member of the Arteiviridae family. Unlike many other enveloped viruses, which possess homotrimeric spikes, the spike responsible for cellular tropism in Arteriviruses is a heterotrimer composed of 3 glycoproteins: GP2, GP3, and GP4. Together with the hydrophobic protein E they are the minor components of virus particles. We describe the expression of all 3 minor glycoproteins, each equipped with a different tag, from a multi-cassette system in mammalian BHK-21 cells. Coprecipitation studies suggest that a rather small faction of GP2, GP3, and GP4 form dimeric or trimeric complexes. GP2, GP3, and GP4 co-localize with each other and also, albeit weaker, with the E-protein. The co-localization of GP3-HA and GP2-myc was tested with markers for ER, ERGIC, and cis-Golgi. The co-localization of GP3-HA was the same regardless of whether it was expressed alone or as a complex, whereas the transport of GP2-myc to cis-Golgi was higher when this protein was expressed as a complex. The glycosylation pattern was also independent of whether the proteins were expressed alone or together. The recombinant spike might be a tool for basic research but might also be used as a subunit vaccine for horses.
Collapse
|
20
|
Wang J, Liu L, Yang D, Zhang L, Abudureyimu A, Qiao Z, Ma Z. Identification and differential expression of microRNAs in Madin–Darby canine kidney cells with high and low tumorigenicities. Genes Genomics 2022; 44:187-196. [DOI: 10.1007/s13258-021-01177-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 10/12/2021] [Indexed: 11/24/2022]
|
21
|
Influenza Vaccine: An Engineering Vision from Virological Importance to Production. BIOTECHNOL BIOPROC E 2022; 27:714-738. [PMID: 36313971 PMCID: PMC9589582 DOI: 10.1007/s12257-022-0115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/06/2022] [Accepted: 06/12/2022] [Indexed: 01/26/2023]
Abstract
According to data from the World Health Organization (WHO) every year, millions of people are affected by flu. Flu is a disease caused by influenza viruses. For preventing this, seasonal influenza vaccinations are widely considered the most efficient way to protect against the negative effects of the flu. To date, there is no "one-size-fits-all" vaccine that can be effective all over the world to protect against all seasonal or pandemic influenza virus types. Because influenza virus transforms its genetic structure and it can emerges as immunogenically new (antigenic drift) which causes epidemics or new virus subtype (antigenic shift) which causes pandemics. As a result, annual revaccination or new subtype viral vaccine development is required. Currently, three types of vaccines (inactivated, live attenuated, and recombinant) are approved in different countries. These can be named "conventional influenza vaccines" and their production are based on eggs or cell culture. Although, there is good effort to develop new influenza vaccines for broader and longer period of time protection. In this sense these candidate vaccines are called "universal influenza vaccines". In this article, after we mentioned the short history of flu then virus morphology and infection, we explained the diseases caused by the influenza virus in humans. Afterward, we explained in detail the production methods of available influenza vaccines, types of bioreactors used in cell culture based production, conventional and new vaccine types, and development strategies for better vaccines.
Collapse
|
22
|
Fulber JPC, Farnós O, Kiesslich S, Yang Z, Dash S, Susta L, Wootton SK, Kamen AA. Process Development for Newcastle Disease Virus-Vectored Vaccines in Serum-Free Vero Cell Suspension Cultures. Vaccines (Basel) 2021; 9:vaccines9111335. [PMID: 34835266 PMCID: PMC8623276 DOI: 10.3390/vaccines9111335] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023] Open
Abstract
The ongoing COVID-19 pandemic drew global attention to infectious diseases, attracting numerous resources for development of pandemic preparedness plans and vaccine platforms—technologies with robust manufacturing processes that can quickly be pivoted to target emerging diseases. Newcastle Disease Virus (NDV) has been studied as a viral vector for human and veterinary vaccines, but its production relies heavily on embryonated chicken eggs, with very few studies producing NDV in cell culture. Here, NDV is produced in suspension Vero cells, and analytical assays (TCID50 and ddPCR) are developed to quantify infectious and total viral titer. NDV-GFP and NDV-FLS (SARS-CoV-2 full-length spike protein) constructs were adapted to replicate in Vero and HEK293 suspension cultures using serum-free media, while fine-tuning parameters such as MOI, temperature, and trypsin concentration. Shake flask productions with Vero cells resulted in infectious titers of 1.07 × 108 TCID50/mL for NDV-GFP and 1.33 × 108 TCID50/mL for NDV-FLS. Production in 1 L batch bioreactors also resulted in high titers in culture supernatants, reaching 2.37 × 108 TCID50/mL for NDV-GFP and 3.16 × 107 TCID50/mL for NDV-FLS. This shows effective NDV production in cell culture, building the basis for a scalable vectored-vaccine manufacturing process that can be applied to different targets.
Collapse
Affiliation(s)
- Julia Puppin Chaves Fulber
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Omar Farnós
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Sascha Kiesslich
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Zeyu Yang
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Shantoshini Dash
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
| | - Leonardo Susta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.S.); (S.K.W.)
| | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.S.); (S.K.W.)
| | - Amine A. Kamen
- Viral Vectors and Vaccines Bioprocessing Group, Department of Bioengineering, McGill University, Montreal, QC H3A 0G4, Canada; (J.P.C.F.); (O.F.); (S.K.); (Z.Y.); (S.D.)
- Correspondence:
| |
Collapse
|
23
|
Cell culture-based production of defective interfering influenza A virus particles in perfusion mode using an alternating tangential flow filtration system. Appl Microbiol Biotechnol 2021; 105:7251-7264. [PMID: 34519855 PMCID: PMC8437742 DOI: 10.1007/s00253-021-11561-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 10/27/2022]
Abstract
Respiratory diseases including influenza A virus (IAV) infections represent a major threat to human health. While the development of a vaccine requires a lot of time, a fast countermeasure could be the use of defective interfering particles (DIPs) for antiviral therapy. IAV DIPs are usually characterized by a large internal deletion in one viral RNA segment. Consequentially, DIPs can only propagate in presence of infectious standard viruses (STVs), compensating the missing gene function. Here, they interfere with and suppress the STV replication and might act "universally" against many IAV subtypes. We recently reported a production system for purely clonal DIPs utilizing genetically modified cells. In the present study, we established an automated perfusion process for production of a DIP, called DI244, using an alternating tangential flow filtration (ATF) system for cell retention. Viable cell concentrations and DIP titers more than 10 times higher than for a previously reported batch cultivation were observed. Furthermore, we investigated a novel tubular cell retention device for its potential for continuous virus harvesting into the permeate. Very comparable performances to typically used hollow fiber membranes were found during the cell growth phase. During the virus replication phase, the tubular membrane, in contrast to the hollow fiber membrane, allowed 100% of the produced virus particles to pass through. To our knowledge, this is the first time a continuous virus harvest was shown for a membrane-based perfusion process. Overall, the process established offers interesting possibilities for advanced process integration strategies for next-generation virus particle and virus vector manufacturing.Key points• An automated perfusion process for production of IAV DIPs was established.• DIP titers of 7.40E + 9 plaque forming units per mL were reached.• A novel tubular cell retention device enabled continuous virus harvesting.
Collapse
|
24
|
Zulkarnain NN, Anuar N, Abd Rahman N, Sheikh Abdullah SR, Alias MN, Yaacob M, Ma Z, Ding G. Cell-based influenza vaccine: current production, halal status assessment, and recommendations towards Islamic-compliant manufacturing. Hum Vaccin Immunother 2021; 17:2158-2168. [PMID: 33539195 DOI: 10.1080/21645515.2020.1865044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Influenza virus is a life-threatening pathogen that infects millions of people every year, with annual mortality in the hundreds of thousands. The scenario for controlling infection has worsened with increasing numbers of vaccine hesitancy cases reported worldwide due to objections on safety, religious and other grounds. Uses of haram (impermissible) and mashbooh (doubtful) ingredients in vaccine production has raised doubts among Muslim consumers and consequently stimulated serious vaccine hesitancy. To address this major problem, we have reviewed and recommended some alternatives appropriate for manufacturing cell-based influenza vaccine which comply with Islamic laws and consumers' needs. Intensive assessments of current influenza vaccine production in both scientific and Islamic views have led to the identification of four main ingredients deemed impermissible in novel sharia-compliant (approved by Islamic laws) vaccine manufacturing. Only some of these impermissible components could be replaced with halal (permissible) alternatives, while others remain impermissible due to unavailability and unsuitability.
Collapse
Affiliation(s)
- Nurul Nadiah Zulkarnain
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Nurina Anuar
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Norliza Abd Rahman
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Siti Rozaimah Sheikh Abdullah
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Muhammad Nazir Alias
- Centre for Contemporary Fiqh and Sharia Compliance, Faculty of Islamic Studies, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Mashitoh Yaacob
- Centre for Liberal Education, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia.,Institute of Islam Hadhari, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Zhongren Ma
- Biomedical Research Centre, Northwest Minzu University, Lanzhou, Gansu, China
| | - Gongtao Ding
- Biomedical Research Centre, Northwest Minzu University, Lanzhou, Gansu, China
| |
Collapse
|
25
|
Silva CAT, Kamen AA, Henry O. Recent advances and current challenges in process intensification of cell culture‐based influenza virus vaccine manufacturing. CAN J CHEM ENG 2021. [DOI: 10.1002/cjce.24197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Cristina A. T. Silva
- Department of Chemical Engineering Polytechnique Montréal Montréal Québec Canada
- Department of Bioengineering McGill University Montréal Québec Canada
| | - Amine A. Kamen
- Department of Bioengineering McGill University Montréal Québec Canada
| | - Olivier Henry
- Department of Chemical Engineering Polytechnique Montréal Montréal Québec Canada
| |
Collapse
|
26
|
Hein MD, Arora P, Marichal-Gallardo P, Winkler M, Genzel Y, Pöhlmann S, Schughart K, Kupke SY, Reichl U. Cell culture-based production and in vivo characterization of purely clonal defective interfering influenza virus particles. BMC Biol 2021; 19:91. [PMID: 33941189 PMCID: PMC8091782 DOI: 10.1186/s12915-021-01020-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 04/01/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Infections with influenza A virus (IAV) cause high morbidity and mortality in humans. Additional to vaccination, antiviral drugs are a treatment option. Besides FDA-approved drugs such as oseltamivir or zanamivir, virus-derived defective interfering (DI) particles (DIPs) are considered promising new agents. IAV DIPs typically contain a large internal deletion in one of their eight genomic viral RNA (vRNA) segments. Consequently, DIPs miss the genetic information necessary for replication and can usually only propagate by co-infection with infectious standard virus (STV), compensating for their defect. In such a co-infection scenario, DIPs interfere with and suppress STV replication, which constitutes their antiviral potential. RESULTS In the present study, we generated a genetically engineered MDCK suspension cell line for production of a purely clonal DIP preparation that has a large deletion in its segment 1 (DI244) and is not contaminated with infectious STV as egg-derived material. First, the impact of the multiplicity of DIP (MODIP) per cell on DI244 yield was investigated in batch cultivations in shake flasks. Here, the highest interfering efficacy was observed for material produced at a MODIP of 1E-2 using an in vitro interference assay. Results of RT-PCR suggested that DI244 material produced was hardly contaminated with other defective particles. Next, the process was successfully transferred to a stirred tank bioreactor (500 mL working volume) with a yield of 6.0E+8 PFU/mL determined in genetically modified adherent MDCK cells. The produced material was purified and concentrated about 40-fold by membrane-based steric exclusion chromatography (SXC). The DI244 yield was 92.3% with a host cell DNA clearance of 97.1% (99.95% with nuclease digestion prior to SXC) and a total protein reduction of 97.2%. Finally, the DIP material was tested in animal experiments in D2(B6).A2G-Mx1r/r mice. Mice infected with a lethal dose of IAV and treated with DIP material showed a reduced body weight loss and all animals survived. CONCLUSION In summary, experiments not only demonstrated that purely clonal influenza virus DIP preparations can be obtained with high titers from animal cell cultures but confirmed the potential of cell culture-derived DIPs as an antiviral agent.
Collapse
Affiliation(s)
- Marc D Hein
- Otto-von-Guericke-University Magdeburg, Chair of Bioprocess Engineering, Magdeburg, Germany
| | - Prerna Arora
- German Primate Center-Leibniz Institute for Primate Research, Infection Biology Unit, Göttingen, Germany.,University Göttingen, Faculty of Biology and Psychology, Göttingen, Germany
| | - Pavel Marichal-Gallardo
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Michael Winkler
- German Primate Center-Leibniz Institute for Primate Research, Infection Biology Unit, Göttingen, Germany.,University Göttingen, Faculty of Biology and Psychology, Göttingen, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Stefan Pöhlmann
- German Primate Center-Leibniz Institute for Primate Research, Infection Biology Unit, Göttingen, Germany.,University Göttingen, Faculty of Biology and Psychology, Göttingen, Germany
| | - Klaus Schughart
- Helmholtz Centre for Infection Research, Department of Infection Genetics, Braunschweig, Germany.,University of Veterinary Medicine Hannover, Hannover, Germany.,University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, TN, USA
| | - Sascha Y Kupke
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany.
| | - Udo Reichl
- Otto-von-Guericke-University Magdeburg, Chair of Bioprocess Engineering, Magdeburg, Germany.,Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| |
Collapse
|
27
|
Shah M, Kumar S. Adaptation and characterization of Anatid herpesvirus 1 in different permissible cell lines. Biologicals 2021; 70:1-6. [PMID: 33744089 DOI: 10.1016/j.biologicals.2021.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/27/2021] [Accepted: 02/21/2021] [Indexed: 11/26/2022] Open
Abstract
Duck viral enteritis is an acute, contagious infection of Anatidae family members. The disease is caused by Anatid herpesvirus 1 (AnHV-1). The infection of AnHV-1 is controlled by vaccination to the flock with chick embryo adapted attenuated vaccine in developed countries. However, its economic impact in developing countries is substantial and there is a need to understand the cell culture spectrum of the virus to produce its vaccine on a mass scale. In the present study, the permissivity of AnHV-1 for different cells was analyzed. The AnHV-1 showed enhanced replication following its serial passage in CEF, DF-1, Vero, MDCK, and QT-35 cells. The characteristic cytopathic effect (CPE) of rounding and clumping of cells were observed in CEF, DF-1, Vero, and QT-35 cell lines. The infectivity and viral replication were highest in CEF, DF-1, Vero, and QT-35 cells. In contrast, the results suggested that MDCK cells are less permissive for AnHV-1 infection with negligible CPE and reduced viral replication. Heterologous cell culture systems other than chicken embryo fibroblasts to adapted live vaccine viruses will provide a system devoid of other avian infectious agents. Moreover, it can be used for the propagation and cultivation of AnHV-1 vaccine strain for developing cell culture-based vaccines with high titer and could be an economical alternative for the existing options.
Collapse
Affiliation(s)
- Manisha Shah
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
28
|
Pralow A, Hoffmann M, Nguyen-Khuong T, Pioch M, Hennig R, Genzel Y, Rapp E, Reichl U. Comprehensive N-glycosylation analysis of the influenza A virus proteins HA and NA from adherent and suspension MDCK cells. FEBS J 2021; 288:4869-4891. [PMID: 33629527 DOI: 10.1111/febs.15787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/04/2021] [Accepted: 02/22/2021] [Indexed: 12/25/2022]
Abstract
Glycosylation is considered as a critical quality attribute for the production of recombinant biopharmaceuticals such as hormones, blood clotting factors, or monoclonal antibodies. In contrast, glycan patterns of immunogenic viral proteins, which differ significantly between the various expression systems, are hardly analyzed yet. The influenza A virus (IAV) proteins hemagglutinin (HA) and neuraminidase (NA) have multiple N-glycosylation sites, and alteration of N-glycan micro- and macroheterogeneity can have strong effects on virulence and immunogenicity. Here, we present a versatile and powerful glycoanalytical workflow that enables a comprehensive N-glycosylation analysis of IAV glycoproteins. We challenged our workflow with IAV (A/PR/8/34 H1N1) propagated in two closely related Madin-Darby canine kidney (MDCK) cell lines, namely an adherent MDCK cell line and its corresponding suspension cell line. As expected, N-glycan patterns of HA and NA from virus particles produced in both MDCK cell lines were similar. Detailed analysis of the HA N-glycan microheterogeneity showed an increasing variability and a higher complexity for N-glycosylation sites located closer to the head region of the molecule. In contrast, NA was found to be exclusively N-glycosylated at site N73. Almost all N-glycan structures were fucosylated. Furthermore, HA and NA N-glycan structures were exclusively hybrid- and complex-type structures, to some extent terminated with alpha-linked galactose(s) but also with blood group H type 2 and blood group A epitopes. In contrast to the similarity of the overall glycan pattern, differences in the relative abundance of individual structures were identified. This concerned, in particular, oligomannose-type, alpha-linked galactose, and multiantennary complex-type N-glycans.
Collapse
Affiliation(s)
- Alexander Pralow
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Marcus Hoffmann
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Terry Nguyen-Khuong
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Markus Pioch
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - René Hennig
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,glyXera GmbH, Magdeburg, Germany
| | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,glyXera GmbH, Magdeburg, Germany
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.,Chair of Bioprocess Engineering, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
29
|
Ganguly M, Yeolekar L, Tyagi P, Sagar U, Narale S, Anaspure Y, Tupe S, Wadkar K, Ingle N, Dhere R, Scorza FB, Mahmood K. Evaluation of manufacturing feasibility and safety of an MDCK cell-based live attenuated influenza vaccine (LAIV) platform. Vaccine 2020; 38:8379-8386. [PMID: 33229107 DOI: 10.1016/j.vaccine.2020.10.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/24/2020] [Accepted: 10/31/2020] [Indexed: 12/23/2022]
Abstract
Cell culture based live attenuated influenza vaccines (LAIV) as an alternative to egg-based LAIV have been explored because of lack of easy access to SPF eggs for large scale production. In this study, feasibility of MDCK platform was assessed by including multiple LAIV strains covering both type A (H1 and H3) and type B seasonal strains as well as the candidate pandemic potential strains like A/H5 and A/H7 for the growth in MDCK cells. A risk assessment study was conducted on the cell banks to evaluate safety concerns related to tumorigenicity with a regulatory perspective. Tumorigenic potential of the MDCK cells was evaluated in nude mice (107cells/mouse) model system. The 50% tumor producing dose (TPD50) of MDCK cells was studied in SCID mice to determine the amount of cells required for induction of tumors. Further, we conducted an oncogenicity study in three sensitive rodent species as per the requirements specified in the WHO guidelines. We determined TPD50 value of 1.9 X 104 cells/mice through subcutaneous route. Our results suggest that, the intranasal route of administration of the cell culture based LAIV pose minimal to no risk of tumorigenicity associated with the host cells. Also, non-oncogenic nature of MDCK cells was demonstrated. Host cell DNA in the vaccine formulations was < 10 ng/dose which ensures vaccine safety. Production efficiency and consistency were characterized and the observed titer values of the viral harvest and the processed bulk were comparable to the expansion in embryonated eggs. The present study clearly establishes the suitability of MDCK cells as a substrate for the manufacture of a safe and viable LAIV.
Collapse
Affiliation(s)
- Milan Ganguly
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India.
| | - Leena Yeolekar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Parikshit Tyagi
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Umesh Sagar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Swapnil Narale
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | | | - Sham Tupe
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Kuntinath Wadkar
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Nilesh Ingle
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | - Rajeev Dhere
- Serum Institute of India Private Limited, 212/2, Hadapsar, Pune, India
| | | | | |
Collapse
|
30
|
Hein MD, Kollmus H, Marichal-Gallardo P, Püttker S, Benndorf D, Genzel Y, Schughart K, Kupke SY, Reichl U. OP7, a novel influenza A virus defective interfering particle: production, purification, and animal experiments demonstrating antiviral potential. Appl Microbiol Biotechnol 2020; 105:129-146. [PMID: 33275160 PMCID: PMC7778630 DOI: 10.1007/s00253-020-11029-5] [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: 09/05/2020] [Revised: 11/14/2020] [Accepted: 11/22/2020] [Indexed: 11/24/2022]
Abstract
Abstract The novel influenza A virus (IAV) defective interfering particle “OP7” inhibits IAV replication in a co-infection and was previously suggested as a promising antiviral agent. Here, we report a batch-mode cell culture-based production process for OP7. In the present study, a seed virus containing standard virus (STV) and OP7 was used. The yield of OP7 strongly depended on the production multiplicity of infection. To inactivate infectious STV in the OP7 material, which may cause harm in a potential application, UV irradiation was used. The efficacy of OP7 in this material was preserved, as shown by an in vitro interference assay. Next, steric exclusion chromatography was used to purify and to concentrate (~ 13-fold) the UV-treated material. Finally, administration of produced OP7 material in mice did not show any toxic effects. Furthermore, all mice infected with a lethal dose of IAV survived the infection upon OP7 co-treatment. Thus, the feasibility of a production workflow for OP7 and its potential for antiviral treatment was demonstrated. Key points • OP7 efficacy strongly depended on the multiplicity of infection used for production • Purification by steric exclusion chromatography increased OP7 efficacy • OP7-treated mice were protected against a lethal infection with IAV Supplementary Information The online version contains supplementary material available at 10.1007/s00253-020-11029-5.
Collapse
Affiliation(s)
- Marc D Hein
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Pavel Marichal-Gallardo
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Sebastian Püttker
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Dirk Benndorf
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,University of Veterinary Medicine Hannover, Hannover, Germany.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Sascha Y Kupke
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.
| | - Udo Reichl
- Bioprocess Engineering, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| |
Collapse
|
31
|
Wu Y, Jia H, Lai H, Liu X, Tan WS. Highly efficient production of an influenza H9N2 vaccine using MDCK suspension cells. BIORESOUR BIOPROCESS 2020. [DOI: 10.1186/s40643-020-00352-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThe use of H9N2 subtype avian influenza vaccines is an effective approach for the control of the virus spread among the poultry, and for the upgrading of vaccine manufacturing, cell culture-based production platform could overcome the limitations of conventional egg-based platform and alternate it. The development of serum-free suspension cell culture could allow even higher virus productivity, where a suspension cell line with good performance and proper culture strategies are required. In this work, an adherent Mardin–Darby canine kidney (MDCK) cell line was adapted to suspension growth to cell concentration up to 12 × 106 cells/mL in a serum-free medium in batch cultures. Subsequently, the H9N2 influenza virus propagation in this MDCK cell line was evaluated with the optimization of infection conditions in terms of MOI and cell concentration for infection. Furthermore, various feed strategies were tested in the infection phase for improved virus titer and a maximum hemagglutinin titer of 13 log2 (HAU/50 μL) was obtained using the 1:2 medium dilution strategy. The evaluation of MDCK cell growth and H9N2 virus production in bioreactors with optimized operating conditions showed comparable cell performance and virus yield compared to shake flasks, with a high cell-specific virus yield above 13,000 virions/cell. With the purified H9N2 virus harvested from the bioreactors, the MDCK cell-derived vaccine was able to induce high titers of neutralizing antibodies in chickens. Overall, the results demonstrate the promising application of the highly efficient MDCK cell-based production platform for the avian influenza vaccine manufacturing.
Collapse
|
32
|
Maegawa K, Sugita S, Arasaki Y, Nerome R, Nerome K. Interleukin 12-containing influenza virus-like-particle vaccine elevate its protective activity against heterotypic influenza virus infection. Heliyon 2020; 6:e04543. [PMID: 32802975 PMCID: PMC7417893 DOI: 10.1016/j.heliyon.2020.e04543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/03/2020] [Accepted: 07/21/2020] [Indexed: 11/20/2022] Open
Abstract
To produce monovalent and bivalent influenza vaccines composed of virus-like particles (VLPs) containing hemagglutinin (HA), we generated four recombinant Baculoviruses derived from Bombyx mori nuclear polyhedrosis virus (BmNPV) and Autographa california nuclear polyhedrosis virus (AcNPV). Monovalent Fukushima (A/tufted duck/Fukushima/16/2011 [H5N1]) (FkH5) and Anhui (A/Anhui/1/2013 [H7N9]) (AnH7) VLP influenza vaccines were produced in silkworm pupae infected with FkH5-BmNPV or AnH7-BmNPV. To produce a bivalent FkH5 and AnH7 vaccine, the pupae were simultaneously inoculated with FkH5-BmNPV and AnH7-BmNPV. Then, interleukin (IL)-containing bivalent vaccines were produced by Eri silkworm pupae following triple infection with FkH5-AcNPV, AnH7-AcNPV, and IL-12-AcNPV. Fluorescent antibody tests in Sf9 cells triple-infected with FkH5-AcNPV, AnH7-AcNPV, and IL-12-AcNPV showed coexpression of FkH5, AnH7, and IL-12 antigens, suggesting the presence of VLPs containing all three antigens. We then performed competitive hemagglutination inhibition (CHI) tests to calculate the VLP vaccine constituents. Inoculation with two recombinant viruses led to the production of bivalent vaccines containing very similar amounts of the H5 and H7 antigens, suggesting that our dual infection system can be used to produce bivalent VLP vaccines. Immunisation of mice with our developed monovalent and bivalent VLP vaccines induced the production of HI antibody, which protected against a sublethal dose of influenza virus. These IL-12-containing vaccines tended to display increased protection against hetero-subtype influenza viruses.
Collapse
Affiliation(s)
- Kenichi Maegawa
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Shigeo Sugita
- Equine Research Institute, Japan Racing Association, 1400-4, Shiba, Shimotsuke-shi, Tochigi 329-0412, Japan
| | - Youta Arasaki
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Reiko Nerome
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Kuniaki Nerome
- The Institute of Biological Resources, 893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
- Corresponding author.
| |
Collapse
|
33
|
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: 61] [Impact Index Per Article: 15.3] [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
|
34
|
Nie J, Sun Y, Peng F, Li X, Yang Y, Liu X, Li Y, Liu C, Bai Z. Production Process Development of Pseudorabies Virus Vaccine by Using a Novel Scale-Down Model of a Fixed-Bed Bioreactor. J Pharm Sci 2019; 109:959-965. [PMID: 31604085 DOI: 10.1016/j.xphs.2019.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/18/2019] [Accepted: 10/01/2019] [Indexed: 12/22/2022]
Abstract
In this study, a novel tube-fixed-bed bioreactor which consists of a TubeSpin bioreactor 50 tube and 0.44 g macrocarriers was developed as the scale-down model of a fixed-bed bioreactor. The adherent Vero cell-based pseudorabies virus (PRV) production process was tested in this novel model. The Vero cells grew well in the tube-fixed-bed bioreactor, and the cell density reached 5.8 × 106 cells/mL after 7 days of culture. The PRV production parameters (time of infection, multiplicity of infection, and harvest process) were optimized in the tube-fixed-bed bioreactor. Then the optimized process (time of infection = 3 days, multiplicity of infection = 0.001 and multiple harvest process) was scaled up 25-fold to an Xcell 1-L laboratory-scale fixed-bed bioreactor and 125-fold to an Xcell 5-L fixed-bed bioreactor successfully. The total PRV harvest in the Xcell 1-L bioreactor at 5 days after infection (dpi) was 10.25 log10 TCID50 which corresponds to 177,827 doses of vaccine. The total PRV harvest in the Xcell 5-L bioreactor at 5 dpi was 11.13 log10 TCID50 which corresponded to 1,348,962 doses of vaccine. The comparable growth curve, metabolism, and PRV production profile of the scaled-up bioreactors confirmed the feasibility and scalability of the tube-fixed-bed bioreactor as a scale-down model of the fixed-bed bioreactor for virus production process development.
Collapse
Affiliation(s)
- Jianqi Nie
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China
| | - Yang Sun
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China
| | - Feng Peng
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China
| | - Xinran Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China
| | - Yankun Yang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China; The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Xiuxia Liu
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Ye Li
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Chunli Liu
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Zhonghu Bai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China; The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
35
|
Lai CC, Weng TC, Tseng YF, Chiang JR, Lee MS, Hu AYC. Evaluation of novel disposable bioreactors on pandemic influenza virus production. PLoS One 2019; 14:e0220803. [PMID: 31404117 PMCID: PMC6690526 DOI: 10.1371/journal.pone.0220803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/23/2019] [Indexed: 01/19/2023] Open
Abstract
Since 1997, the highly pathogenic influenza H5N1 virus has spread from Hong Kong. According to the WHO bulletin report, the H5N1 virus is a zoonotic disease threat that has infected more than 850 humans, causing over 450 deaths. In addition, an outbreak of another new and highly pathogenic influenza virus (H7N9) occurred in 2013 in China. These highly pathogenic influenza viruses could potentially cause a worldwide pandemic. it is crucial to develop a rapid production platform to meet this surge demand against any possible influenza pandemic. A potential solution for this problem is the use of cell-based bioreactors for rapid vaccine production. These novel bioreactors, used for cell-based vaccine production, possess various advantages. For example, they enable a short production time, allow for the handling highly pathogenic influenza in closed environments, and can be easily scaled up. In this study, two novel disposable cell-based bioreactors, BelloCell and TideCell, were used to produce H5N1 clade II and H7N9 candidate vaccine viruses (CVVs). Madin-Darby canine kidney (MDCK) cells were used for the production of these influenza CVVs. A novel bench-scale bioreactor named BelloCell bioreactor was used in the study. All culturing conditions were tested and scaled to 10 L industrial-scale bioreactor known as TideCell002. The performances of between BelloCell and TideCell were similar in cell growth, the average MDCK cell doubling time was slightly decreased to 25 hours. The systems yielded approximately 39.2 and 18.0 μg/ml of HA protein with the 10-liter TideCell002 from the H5N1 clade II and H7N9 CVVs, respectively. The results of this study not only highlight the overall effectiveness of these bioreactors but also illustrate the potential of maintaining the same outcome when scaled up to industrial production, which has many implications for faster vaccine production. Although additional studies are required for process optimization, the results of this study are promising and show that oscillating bioreactors may be a suitable platform for pandemic influenza virus production.
Collapse
Affiliation(s)
- Chia-Chun Lai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- College of Life Science Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Tsai-Chuan Weng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Fen Tseng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Jen-Ron Chiang
- Vaccine Center, Centers for Disease Control, Taipei, Taiwan
| | - Min-Shi Lee
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Alan Yung-Chih Hu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- * E-mail:
| |
Collapse
|
36
|
Gränicher G, Coronel J, Pralow A, Marichal-Gallardo P, Wolff M, Rapp E, Karlas A, Sandig V, Genzel Y, Reichl U. Efficient influenza A virus production in high cell density using the novel porcine suspension cell line PBG.PK2.1. Vaccine 2019; 37:7019-7028. [PMID: 31005427 DOI: 10.1016/j.vaccine.2019.04.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 12/22/2022]
Abstract
Seasonal and pandemic influenza respiratory infections are still a major public health issue. Vaccination is the most efficient way to prevent influenza infection. One option to produce influenza vaccines is cell-culture based virus propagation. Different host cell lines, such as MDCK, Vero, AGE1.CR or PER.C6 cells have been shown to be a good substrate for influenza virus production. With respect to the ease of scale-up, suspension cells should be preferred over adherent cells. Ideally, they should replicate different influenza virus strains with high cell-specific yields. Evaluation of new cell lines and further development of processes is of considerable interest, as this increases the number of options regarding the design of manufacturing processes, flexibility of vaccine production and efficiency. Here, PBG.PK2.1, a new mammalian cell line that was developed by ProBioGen AG (Germany) for virus production is presented. The cells derived from immortal porcine kidney cells were previously adapted to growth in suspension in a chemically-defined medium. Influenza virus production was improved after virus adaptation to PBG.PK2.1 cells and optimization of infection conditions, namely multiplicity of infection and trypsin concentration. Hemagglutinin titers up to 3.24 log10(HA units/100 µL) were obtained in fed-batch mode in bioreactors (700 mL working volume). Evaluation of virus propagation in high cell density culture using a hollow-fiber based system (ATF2) demonstrated promising performance: Cell concentrations of up to 50 × 106 cells/mL with viabilities exceeding 95%, and a maximum HA titer of 3.93 log10(HA units/100 µL). Analysis of glycosylation of the viral HA antigen expressed showed clear differences compared to HA produced in MDCK or Vero cell lines. With an average cell-specific productivity of 5000 virions/cell, we believe that PBG.PK2.1 cells are a very promising candidate to be considered for next-generation influenza virus vaccine production.
Collapse
Affiliation(s)
- Gwendal Gränicher
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany.
| | - Juliana Coronel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany
| | - Alexander Pralow
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany
| | - Pavel Marichal-Gallardo
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany
| | - Michael Wolff
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany; Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390 Gießen, Germany
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany
| | | | | | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Sandtorstr. 1, 39106 Magdeburg, Germany; Chair for Bioprocess Engineering, Otto-von-Guericke-University Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
| |
Collapse
|
37
|
Durous L, Julien T, Padey B, Traversier A, Rosa-Calatrava M, Blum LJ, Marquette CA, Petiot E. SPRi-based hemagglutinin quantitative assay for influenza vaccine production monitoring. Vaccine 2019; 37:1614-1621. [DOI: 10.1016/j.vaccine.2019.01.083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/15/2019] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
|
38
|
Tsai HC, Lehman CW, Lin CC, Tsai SW, Chen CM. Functional evaluation for adequacy of MDCK-lineage cells in influenza research. BMC Res Notes 2019; 12:101. [PMID: 30808400 PMCID: PMC6390338 DOI: 10.1186/s13104-019-4134-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/15/2019] [Indexed: 11/23/2022] Open
Abstract
Objective Influenza is an acute respiratory disease caused by the influenza virus which circulates annually in populations of different species. Madin-Darby Canine Kidney (MDCK) is the most widely utilized cell-line for conducting influenza research. However, the infectivity of various influenza strains in MDCK cells is not equivalent and the productivity of viral propagation is also limited. Results We tested the functional adequacy of two MDCK-lineage cell lines, conventional MDCK and MDCK/London, were evaluated by assessing their infectivity of different influenza viral strains with focus forming assays and the cellular toxicity caused by influenza infections by lactate dehydrogenase assay. Moreover, the sensitivity of cells in the presence of the antiviral agent ribavirin was assessed by MTT assay. Our results showed that MDCK/London cells efficiently propagate virus across all influenza viruses tested, are comparable to the utility of Mv1Lu cells, and are superior to conventional MDCK cells in replicating virus as indicated by an increase in virus of three to four logs, particularly in H3N2 infection. Also, the MDCK/London cells were more sensitive to the presence of antiviral drug than conventional MDCK cells. In conclusion, MDCK/London cell line could be a better platform for influenza studies and vaccine development.
Collapse
Affiliation(s)
- Hsin-Chung Tsai
- Department of Surgery, Taichung Hospital, Ministry of Health and Welfare, 199 San Min Rd., Sec. 1, Taichung, Taiwan. .,Department of Life Sciences, The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xinda Rd., Taichung, 402, Taiwan.
| | - Caitlin W Lehman
- School of System Biology, George Mason University, Manassas, VA, USA
| | - Chi-Chieh Lin
- Department of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Sen-Wei Tsai
- Department of Physical Medicine and Rehabilitation, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, The iEGG and Animal Biotechnology Center, National Chung Hsing University, 145 Xinda Rd., Taichung, 402, Taiwan.
| |
Collapse
|
39
|
Looi QH, Foo JB, Lim MT, Le CF, Show PL. How far have we reached in development of effective influenza vaccine? Int Rev Immunol 2018; 37:266-276. [PMID: 30252547 DOI: 10.1080/08830185.2018.1500570] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite of ongoing research programs and numerous clinical trials, seasonal influenza epidemics remain a major concern globally. Vaccination remains the most effective method to prevent influenza infection. However, current flu vaccines have several limitations, including limited vaccine capacity, long production times, inconsistence efficacy in certain populations, and lack of a "universal" solution. Different next-generation approaches such as cell line-based culture, reverse genetics, and virus expression technology are currently under development to address the aforementioned challenges in conventional vaccine manufacture pipeline. Such approaches hope for safe and scalable production, induce broad-spectrum immunity, create premade libraries of vaccine strains, and target nonvariable regions of antigenic proteins for "universal" vaccination. Here, we discuss the process and challenges of the current influenza vaccine platform as well as new approaches that are being investigated. These developments indicate that an exciting future lies ahead in the influenza vaccine field.
Collapse
Affiliation(s)
- Qi Hao Looi
- a Ming Medical Services Sdn. Bhd , Petaling Jaya , Selangor Darul Ehsan , Malaysia
| | - Jhi Biau Foo
- b School of Pharmacy, Faculty of Health & Medical Sciences , Taylor's University , Subang Jaya , Selangor Darul Ehsan , Malaysia
| | - May Teng Lim
- c Department of Chemical and Environmental Engineering, Faculty of Engineering , University of Nottingham Malaysia Campus , Jalan Braga , Semenyih, Selangor Darul Ehsan , Malaysia
| | - Cheng Foh Le
- d School of Biosciences, Faculty of Science , University of Nottingham Malaysia Campus , Jalan Broga , Semenyih , Selangor Darul Ehsan , Malaysia
| | - Pau Loke Show
- c Department of Chemical and Environmental Engineering, Faculty of Engineering , University of Nottingham Malaysia Campus , Jalan Braga , Semenyih, Selangor Darul Ehsan , Malaysia.,e Molecular Pharming and Bioproduction Research Group, Food and Pharmaceutical Engineering Research Group, Department of Chemical and Environmental Engineering, Faculty of Engineering , University of Nottingham Malaysia Campus , Jalan Broga, Semenyih , Selangor Darul Ehsan , Malaysia
| |
Collapse
|
40
|
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: 6.7] [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
|
41
|
Wang H, Guo S, Li Z, Xu X, Shao Z, Song G. Suspension culture process for H9N2 avian influenza virus (strain Re-2). Arch Virol 2017; 162:3051-3059. [DOI: 10.1007/s00705-017-3460-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 05/20/2017] [Indexed: 01/18/2023]
|
42
|
Influenza viruses production: Evaluation of a novel avian cell line DuckCelt®-T17. Vaccine 2017; 36:3101-3111. [PMID: 28571695 DOI: 10.1016/j.vaccine.2017.03.102] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 03/17/2017] [Accepted: 03/31/2017] [Indexed: 12/22/2022]
Abstract
The influenza vaccine manufacturing industry is looking for production cell lines that are easily scalable, highly permissive to multiple viruses, and more effective in term of viral productivity. One critical characteristic of such cell lines is their ability to grow in suspension, in serum free conditions and at high cell densities. Influenza virus causing severe epidemics both in human and animals is an important threat to world healthcare. The repetitive apparition of influenza pandemic outbreaks in the last 20years explains that manufacturing sector is still looking for more effective production processes to replace/supplement embryonated egg-based process. Cell-based production strategy, with a focus on avian cell lines, is one of the promising solutions. Three avian cell lines, namely duck EB66®cells (Valneva), duck AGE.CR® cells (Probiogen) and quail QOR/2E11 cells (Baxter), are now competing with traditional mammalian cell platforms (Vero and MDCK cells) used for influenza vaccine productions and are currently at advance stage of commercial development for the manufacture of influenza vaccines. The DuckCelt®-T17 cell line presented in this work is a novel avian cell line developed by Transgene. This cell line was generated from primary embryo duck cells with the constitutive expression of the duck telomerase reverse transcriptase (dTERT). The DuckCelt®-T17 cells were able to grow in batch suspension cultures and serum-free conditions up to 6.5×106cell/ml and were easily scaled from 10ml up to 3l bioreactor. In the present study, DuckCelt®-T17 cell line was tested for its abilities to produce various human, avian and porcine influenza strains. Most of the viral strains were produced at significant infectious titers (>5.8 log TCID50/ml) with optimization of the infection conditions. Human strains H1N1 and H3N2, as well as all the avian strains tested (H5N2, H7N1, H3N8, H11N9, H12N5) were the most efficiently produced with highest titre reached of 9.05 log TCID50/ml for A/Panama/2007/99 influenza H3N2. Porcine strains were also greatly rescued with titres from 4 to 7 log TCID50/ml depending of the subtypes. Interestingly, viral kinetics showed maximal titers reached at 24h post-infection for most of the strains, allowing early harvest time (Time Of Harvest: TOH). The B strains present specific production kinetics with a delay of 24h before reaching the maximal viral particle release. Process optimization on H1N1 2009 human pandemic strain allowed identifying best operating conditions for production (MOI, trypsin concentration, cell density at infection) allowing improving the production level by 2 log. Our results suggest that the DuckCelt®-T17 cell line is a very promising platform for industrial production of influenza viruses and particularly for avian viral strains.
Collapse
|
43
|
Porcine Sapelovirus Uses α2,3-Linked Sialic Acid on GD1a Ganglioside as a Receptor. J Virol 2016; 90:4067-4077. [PMID: 26865725 PMCID: PMC4810533 DOI: 10.1128/jvi.02449-15] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/14/2016] [Indexed: 12/28/2022] Open
Abstract
The receptor(s) for porcine sapelovirus (PSV), which causes diarrhea, pneumonia, polioencephalomyelitis, and reproductive disorders in pigs, remains largely unknown. Given the precedent for other picornaviruses which use terminal sialic acids (SAs) as receptors, we examined the role of SAs in PSV binding and infection. Using a variety of approaches, including treating cells with a carbohydrate-destroying chemical (NaIO4), mono- or oligosaccharides (N-acetylneuraminic acid, galactose, and 6′-sialyllactose), linkage-specific sialidases (neuraminidase and sialidase S), lectins (Maakia amurensis lectin and Sambucus nigra lectin), proteases (trypsin and chymotrypsin), and glucosylceramide synthase inhibitors (dl-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol and phospholipase C), we demonstrated that PSV could recognize α2,3-linked SA on glycolipids as a receptor. On the other hand, PSVs had no binding affinity for synthetic histo-blood group antigens (HBGAs), suggesting that PSVs could not use HBGAs as receptors. Depletion of cell surface glycolipids followed by reconstitution studies indicated that GD1a ganglioside, but not other gangliosides, could restore PSV binding and infection, further confirming α2,3-linked SA on GD1a as a PSV receptor. Our results could provide significant information on the understanding of the life cycle of sapelovirus and other picornaviruses. For the broader community in the area of pathogens and pathogenesis, these findings and insights could contribute to the development of affordable, useful, and efficient drugs for anti-sapelovirus therapy. IMPORTANCE The porcine sapelovirus (PSV) is known to cause enteritis, pneumonia, polioencephalomyelitis, and reproductive disorders in pigs. However, the receptor(s) that the PSV utilizes to enter host cells remains largely unknown. Using a variety of approaches, we showed that α2,3-linked terminal sialic acid (SA) on the cell surface GD1a ganglioside could be used for PSV binding and infection as a receptor. On the other hand, histo-blood group antigens also present in the cell surface carbohydrates could not be utilized as PSV receptors for binding and infection. These findings should contribute to the understanding of the sapelovirus life cycle and to the development of affordable, useful and efficient drugs for anti-sapelovirus therapy.
Collapse
|
44
|
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: 6.3] [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
|
45
|
Preparation of microcarriers based on zein and their application in cell culture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 58:863-9. [DOI: 10.1016/j.msec.2015.09.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 08/12/2015] [Accepted: 09/10/2015] [Indexed: 11/22/2022]
|
46
|
Huang D, Peng WJ, Ye Q, Liu XP, Zhao L, Fan L, Xia-Hou K, Jia HJ, Luo J, Zhou LT, Li BB, Wang SL, Xu WT, Chen Z, Tan WS. Serum-Free Suspension Culture of MDCK Cells for Production of Influenza H1N1 Vaccines. PLoS One 2015; 10:e0141686. [PMID: 26540170 PMCID: PMC4634975 DOI: 10.1371/journal.pone.0141686] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 10/12/2015] [Indexed: 01/03/2023] Open
Abstract
Development of serum-free suspension cell culture processes is very important for influenza vaccine production. Previously, we developed a MDCK suspension cell line in a serum-free medium. In the present study, the growth kinetics of suspension MDCK cells and influenza virus production in the serum-free medium were investigated, in comparison with those of adherent MDCK cells in both serum-containing and serum-free medium. It was found that the serum-free medium supported the stable subculture and growth of both adherent and suspension cells. In batch culture, for both cell lines, the growth kinetics in the serum-free medium was comparable with those in the serum-containing medium and a commercialized serum-free medium. In the serum-free medium, peak viable cell density (VCD), haemagglutinin (HA) and median tissue culture infective dose (TCID50) titers of the two cell lines reached 4.51×106 cells/mL, 2.94Log10(HAU/50 μL) and 8.49Log10(virions/mL), and 5.97×106 cells/mL, 3.88Log10(HAU/50 μL), and 10.34Log10(virions/mL), respectively. While virus yield of adherent cells in the serum-free medium was similar to that in the serum-containing medium, suspension culture in the serum-free medium showed a higher virus yield than adherent cells in the serum-containing medium and suspension cells in the commercialized serum-free medium. However, the percentage of infectious viruses was lower for suspension culture in the serum-free medium. These results demonstrate the great potential of this suspension MDCK cell line in serum-free medium for influenza vaccine production and further improvements are warranted.
Collapse
Affiliation(s)
- Ding Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wen-Juan Peng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qian Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xu-Ping Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- * E-mail: (X-PL); (W-ST)
| | - Liang Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Li Fan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Kang Xia-Hou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Han-Jing Jia
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jian Luo
- Shanghai Institute of Biological Products Co., Ltd., Shanghai 200052, China
| | - Lin-Ting Zhou
- Shanghai Institute of Biological Products Co., Ltd., Shanghai 200052, China
| | - Bei-Bei Li
- Shanghai Institute of Biological Products Co., Ltd., Shanghai 200052, China
| | - Shi-Lei Wang
- Shanghai Institute of Biological Products Co., Ltd., Shanghai 200052, China
| | - Wen-Ting Xu
- Shanghai Institute of Biological Products Co., Ltd., Shanghai 200052, China
| | - Ze Chen
- Shanghai Institute of Biological Products Co., Ltd., Shanghai 200052, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- * E-mail: (X-PL); (W-ST)
| |
Collapse
|
47
|
Mattos DA, Silva MV, Gaspar LP, Castilho LR. Increasing Vero viable cell densities for yellow fever virus production in stirred-tank bioreactors using serum-free medium. Vaccine 2015; 33:4288-91. [DOI: 10.1016/j.vaccine.2015.04.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 03/30/2015] [Accepted: 04/02/2015] [Indexed: 10/23/2022]
|
48
|
Soema PC, Kompier R, Amorij JP, Kersten GFA. Current and next generation influenza vaccines: Formulation and production strategies. Eur J Pharm Biopharm 2015; 94:251-63. [PMID: 26047796 DOI: 10.1016/j.ejpb.2015.05.023] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
Abstract
Vaccination is the most effective method to prevent influenza infection. However, current influenza vaccines have several limitations. Relatively long production times, limited vaccine capacity, moderate efficacy in certain populations and lack of cross-reactivity are important issues that need to be addressed. We give an overview of the current status and novel developments in the landscape of influenza vaccines from an interdisciplinary point of view. The feasibility of novel vaccine concepts not only depends on immunological or clinical outcomes, but also depends on biotechnological aspects, such as formulation and production methods, which are frequently overlooked. Furthermore, the next generation of influenza vaccines is addressed, which hopefully will bring cross-reactive influenza vaccines. These developments indicate that an exciting future lies ahead in the influenza vaccine field.
Collapse
Affiliation(s)
- Peter C Soema
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands; Division of Drug Delivery and Technology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| | - Ronald Kompier
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands; FluConsult, Noordwijk, The Netherlands
| | - Jean-Pierre Amorij
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands.
| | - Gideon F A Kersten
- Intravacc (Institute for Translational Vaccinology), Bilthoven, The Netherlands; Division of Drug Delivery and Technology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands
| |
Collapse
|
49
|
Zhang H, Han Q, Ping X, Li L, Chang C, Chen Z, Shu Y, Xu K, Sun B. A single NS2 mutation of K86R promotes PR8 vaccine donor virus growth in Vero cells. Virology 2015; 482:32-40. [PMID: 25817403 DOI: 10.1016/j.virol.2015.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 12/30/2014] [Accepted: 03/02/2015] [Indexed: 02/05/2023]
Abstract
Vaccination is the most effective way to prevent and control infection by influenza viruses, and a cell-culture-based vaccine production system is preferred as the future choice for the large-scale production of influenza vaccines. As one of the WHO-recommended cell lines for producing influenza vaccines, Vero cells do not efficiently support the growth of the current influenza A virus vaccine donor strain, the A/Puerto Rico/8/1934 (PR8) virus. In this study, a single mutation of K86R in the NS2 protein can sufficiently render the high-yielding property to the PR8 virus in Vero cells. Further analysis showed that the later steps in the virus replication cycle were accelerated by NS2(K86R) mutation, which may relate to an enhanced interaction between NS2(K86R) and the components of host factor F1Fo-ATPase, FoB and F1β. Because the NS2(K86R) mutation does not increase PR8 virulence in either mice or embryonated eggs, the PR8-NS2(K86R) virus could serve as a promising vaccine donor strain in Vero cells.
Collapse
Affiliation(s)
- Hong Zhang
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Qinglin Han
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Xianqiang Ping
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China; Shanghai Normal University, No. 100 Guilin Road, Shanghai 200234, China
| | - Li Li
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Chong Chang
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | - Ze Chen
- Shanghai Institute of Biological Products, Shanghai 200052, China
| | - Yuelong Shu
- Chinese Center for Disease Control and Prevention, Yingxin Street 100, Xuanwu District, Beijing 100052, China
| | - Ke Xu
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China.
| | - Bing Sun
- Key Laboratory of Molecular Virology & Immunology, Institute Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China; State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China.
| |
Collapse
|
50
|
Kaiser SC, Kraume M, Eibl D, Eibl R. Single-Use Bioreactors for Animal and Human Cells. CELL ENGINEERING 2015. [DOI: 10.1007/978-3-319-10320-4_14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|