1
|
Nguyen TNT, Park D, Canova CT, Sangerman J, Srinivasan P, Ou RW, Barone PW, Neufeld C, Wolfrum JM, Springs SL, Sinskey AJ, Braatz RD. Perfusion-Based Production of rAAV via an Intensified Transient Transfection Process. Biotechnol Bioeng 2025; 122:1424-1440. [PMID: 40103325 PMCID: PMC12067042 DOI: 10.1002/bit.28967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 01/05/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025]
Abstract
Increasing demand for recombinant adeno-associated virus (rAAV)-based gene therapies necessitates increased manufacturing production. Transient transfection of mammalian cells remains the most commonly used method to produce clinical-grade rAAVs due to its ease of implementation. However, transient transfection processes are often characterized by suboptimal yields and low fractions of full-to-total capsids, both of which contribute to the high cost of goods of many rAAV-based gene therapies. Our previously developed mechanistic model for rAAV2/5 production indicated that the inadequate capsid filling is due to a temporal misalignment between viral DNA replication and capsid synthesis within the cells and the repression of later phase capsid formation by Rep proteins. We experimentally validated this prediction and showed that performing multiple, time-separated doses of plasmid increases the production of rAAV. In this study, we use the insights generated by our mechanistic model to develop an intensified process for rAAV production that combines perfusion with high cell density re-transfection. We demonstrate that performing multiple, time-separated doses at high cell density boosts both cell-specific and volumetric productivity and improves plasmid utilization when compared to a single bolus at standard operating conditions. Our results establish a new paradigm for continuously manufacturing rAAV via transient transfection that improves productivity and reduces manufacturing costs.
Collapse
Affiliation(s)
- Tam N. T. Nguyen
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Damdae Park
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Christopher T. Canova
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Jose Sangerman
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Prasanna Srinivasan
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Rui Wen Ou
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Paul W. Barone
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Caleb Neufeld
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Jacqueline M. Wolfrum
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Stacy L. Springs
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Anthony J. Sinskey
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Richard D. Braatz
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Center for Biomedical InnovationMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| |
Collapse
|
2
|
Kuo HJ, Srinivasan P, Lin YC, Lu M, Rungkittikhun C, Zhang Q, Hu WS. Transcriptomic functional characterization of recombinant adeno-associated virus producing cell line adapted to suspension-growth. Biotechnol Prog 2025:e70042. [PMID: 40396307 DOI: 10.1002/btpr.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/23/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Recombinant adeno-associated virus (rAAV) is a widely used delivery vehicle in gene therapy. A scalable production technology is essential for its wide clinical applications. We have taken a synthetic biology approach to generate HEK293-based cell lines which harbor integrated genetic elements encoding essential AAV and adenoviral helper components and can be induced to produce rAAV. Through cycles of cell line enhancement, a high rAAV productivity could be achieved. The cell lines, like their parental HEK293, grew adherently. For scalable production, cell cultivation in suspension is highly desirable. A producer cell line GX6B was adapted to suspension growth in serum-free medium (named GX6Bs). However, it had substantially reduced virus titer. Returning GX6Bs cells to adherent culture conditions using adherent medium and cultured stationarily brought the productivity back to close to the level of adherent GX6B. A survey of the transcriptome revealed that induction and rAAV production elicited a wide range of cellular changes in various functional classes, including host immune defense response and nucleosome organization. The response was more subdued in suspension-growing GX6Bs. Upon reverting to adherent growth, the cellular transcriptome change regained its vigor to be more similar to that seen in GX6B. The GX6Bs maintained in suspension serum-free conditions were then reverted to the adherent culture medium but under an agitated culture environment to keep suspension growth for rAAV production. The productivity returned to within 25%-50% of GX6B. This work demonstrated the feasibility of the suspension culture of synthetic cell lines for the expansion and production of rAAV.
Collapse
Affiliation(s)
- Han-Jung Kuo
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Prahalad Srinivasan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yu-Chieh Lin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Min Lu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Carissa Rungkittikhun
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qi Zhang
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Nie J, Xu Z, Sun Y, Ren H, Song Z, Zhang Y, Bai Z. Improved productivity of recombinant adeno-associated virus (rAAV) via triple transfection of HEK293 cells using perfusion cultivation. Bioprocess Biosyst Eng 2025:10.1007/s00449-025-03167-9. [PMID: 40285883 DOI: 10.1007/s00449-025-03167-9] [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: 08/18/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
In recent years, recombinant adeno-associated virus (rAAV) vectors are the promising viral transfer tools for gene therapy and clinical trials, thanks to their favorable safety profile and long-term transgene expression. The increasing demand for rAAVs for gene therapy led to a rise in the amount of these vectors required for pre-clinical trials, clinical trials, and approved therapeutic applications. A majority of suspension HEK293 cell-based rAAV production protocols reported rely on a triple transfection at cell density below 2 × 106 cells/mL. However, the low yield of such biomanufacturing challenges bioprocess engineers to develop more efficient strategies capable of increasing volumetric productivity. In this study, we developed a perfusion bioprocess to enable rAAV production efficiently at high cell density. We first optimized three key process parameters (the total DNA amount, ratio of polyethyleneimine (PEI) to DNA, and proportion of the three plasmids) of rAAV production at cell density of 2 × 106 cells/mL by the design of experiment method, from which the robust setpoint (total DNA amount of 1.37 μg/mL, ratio of PEI to DNA of 1.52 μL/μg, the proportion of plasmids pHelper 24%, pRC 46%, pGOI 30%) was explored. We then developed a rAAV production process at a cell density of ~ 8 × 106 cells/mL, with increasing DNA amount on a cell basis and optimizing transfection complex preparation. This approach was confirmed in a 5 L benchtop bioreactor connected with a perfusion system, resulting in a viral genomic titer of 7.28 × 1011 vg/mL and a cell-specific viral genomic titer of 4.97 × 104 vg/cell. This study demonstrates that the perfusion process coupled with optimized transfection complex preparation has the potential to improve manufacturing productivity.
Collapse
Affiliation(s)
- Jianqi Nie
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Zhaojing Xu
- Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yang Sun
- School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - He Ren
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214122, China
| | - Zichuan Song
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, 214122, China
| | - Yan Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China.
| | - Zhonghu Bai
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, 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.
- Zhengzhou University of Technology, Zhengzhou, 450044, China.
| |
Collapse
|
4
|
Zehetner L, Széliová D, Kraus B, Hernandez Bort JA, Zanghellini J. Multi-omics driven genome-scale metabolic modeling improves viral vector yield in HEK293. Metab Eng 2025; 91:103-118. [PMID: 40220853 DOI: 10.1016/j.ymben.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/06/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
HEK293 cells are a versatile cell line extensively used in the production of recombinant proteins and viral vectors, notably Adeno-associated virus (AAV) (Bulcha et al., 2021). Despite their high transfection efficiency and adaptability to various culture conditions, challenges remain in achieving sufficient yields of active viral particles. This study presents a comprehensive multi-omics analysis of two HEK293 strains under good manufacturing practice conditions, focusing on the metabolic and cellular responses during AAV production. The investigation included lipidomic, exometabolomic, and transcriptomic profiling across different conditions and time points. Genome-scale metabolic models (GSMMs) were reconstructed for these strains to elucidate metabolic shifts and identify potential bottlenecks in AAV production. Notably, the study revealed significant differences between a High-producing (HP) and a Low-producing (LP) HEK293 strains, highlighting pseudohypoxia in the LP strain. Key findings include the identification of hypoxia-inducible factor 1-alpha (HIF-1α) as a critical regulator in the LP strain, linking pseudohypoxia to poor AAV productivity. Inhibition of HIF-1α resulted in immediate cessation of cell growth and a 2.5-fold increase in viral capsid production, albeit with a decreased number of viral genomes, impacting the full-to-empty particle ratio. This trade-off is significant because it highlights a key challenge in AAV production: achieving a balance between capsid assembly and genome packaging to optimize the yield of functional viral vectors. Overall this suggests that while HIF-1α inhibition enhances capsid assembly, it simultaneously hampers nucleotide synthesis via the pentose phosphate pathway (PPP), necessary for nucleotide synthesis, and therefore for AAV genome replication.
Collapse
Affiliation(s)
- L Zehetner
- Department for Analytical Chemistry, University of Vienna, Vienna, 1090, Austria; Doctoral School of Chemistry, University of Vienna, Vienna, 1090, Austria.
| | - D Széliová
- Department for Analytical Chemistry, University of Vienna, Vienna, 1090, Austria.
| | - B Kraus
- Institute of Molecular Biotechnology, Institut für Molekulare Biotechnologie GmbH, Vienna, 1030, Austria
| | - J A Hernandez Bort
- Department of Applied Life Sciences, Bioengineering, University of Applied Sciences Campus Vienna, Vienna, 1100, Austria.
| | - J Zanghellini
- Department for Analytical Chemistry, University of Vienna, Vienna, 1090, Austria.
| |
Collapse
|
5
|
Green EA, Fu Q, Ndhairo N, Leibiger TM, Wang Y, Lee Y, Lee KH, Betenbaugh M, Yoon S, McNally DJ. Development of an HEK293 Suspension Cell Culture Medium, Transient Transfection Optimization Workflow, and Analytics for Batch rAAV Manufacturing. Biotechnol Bioeng 2025. [PMID: 40197832 DOI: 10.1002/bit.28980] [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: 12/18/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025]
Abstract
Recombinant adeno associated virus (rAAV) vectors have become popular delivery vehicles for in vivo gene therapies, but demand for rAAVs continues to outpace supply. Platform processes for rAAV production are being developed by many manufacturers, and transient chemical transfection of human embryonic kidney 293 (HEK293) cells is currently the most popular approach. However, the cutting edge nature of rAAV process development encourages manufacturers to keep cell culture media formulations, plasmid sequences, and other details proprietary, which creates hurdles for small companies and academic labs seeking to innovate in this space. To address this problem, we leveraged the resources of an academic-industry consortium (Advanced Mammalian Biomanufacturing Innovation Center, AMBIC) to develop an rAAV production system based on transient transfection of suspension HEK293 cells adapted to an in-house, chemically defined medium. We found that balancing iron and calcium levels in the medium were crucial for maintaining transfection efficiency and minimizing cell aggregation, respectively. A design of experiments approach was used to optimize the transient transfection process for batch rAAV production, and PEI:DNA ratio and cell density at transfection were the parameters with the strongest effects on vector genome (VG) titer. When the optimized transient process was transferred between two university sites, VG titers were within a twofold range. Analytical characterization showed that purified rAAV from the AMBIC process had comparable viral protein molecular weights versus vector derived from commercial processes, but differences in transducing unit (TU) titer were observed between vector preps. The developed media formulation, transient transfection process, and analytics for VG titer, capsid identity, and TU titer constitute a set of workflows that can be adopted by others to study fundamental problems that could improve product yield and quality in the nascent field of rAAV manufacturing.
Collapse
Affiliation(s)
- Erica A Green
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Qiang Fu
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Nelson Ndhairo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Thomas M Leibiger
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Yongdan Wang
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Yongsuk Lee
- Department of Pharmaceutical Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Kelvin H Lee
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA
| | - Michael Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Seongkyu Yoon
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - David J McNally
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
- MassBiologics, University of Massachusetts Chan Medical School, Fall River, Massachusetts, USA
| |
Collapse
|
6
|
Zwi-Dantsis L, Mohamed S, Massaro G, Moeendarbary E. Adeno-Associated Virus Vectors: Principles, Practices, and Prospects in Gene Therapy. Viruses 2025; 17:239. [PMID: 40006994 PMCID: PMC11861813 DOI: 10.3390/v17020239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Gene therapy offers promising potential as an efficacious and long-lasting therapeutic option for genetic conditions, by correcting defective mutations using engineered vectors to deliver genetic material to host cells. Among these vectors, adeno-associated viruses (AAVs) stand out for their efficiency, versatility, and safety, making them one of the leading platforms in gene therapy. The enormous potential of AAVs has been demonstrated through their use in over 225 clinical trials and the FDA's approval of six AAV-based gene therapy products, positioning these vectors at the forefront of the field. This review highlights the evolution and current applications of AAVs in gene therapy, focusing on their clinical successes, ongoing developments, and the manufacturing processes required for the rapid commercial growth anticipated in the AAV therapy market. It also discusses the broader implications of these advancements for future therapeutic strategies targeting more complex and multi-systemic conditions and biological processes such as aging. Finally, we explore some of the major challenges currently confronting the field.
Collapse
Affiliation(s)
- Limor Zwi-Dantsis
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Saira Mohamed
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| |
Collapse
|
7
|
Deng Z, Lv YL, Wang XT, Yuan LH, Zhao K, Du ZM, Xiao X. Production of Recombinant Adeno-Associated Virus Through High-Cell-Density Transfection of HEK293 Cells Based on Fed-Perfusion Culture. Hum Gene Ther 2025; 36:116-127. [PMID: 39761134 DOI: 10.1089/hum.2024.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Adeno-associated virus (AAV)-associated gene therapy has been increasingly promising, in light of the drugs progressed to clinical trials or approved for medications internationally. Therefore, scalable and efficient production of recombinant AAV is pivotal for advancing gene therapy. Traditional methods, such as the triple-plasmid transfection of human embryonic kidney 293 cells in suspension culture, have been widely employed but often hampered by low unit yield. In this study, we optimized the cell culture process with high cell density up to 2 × 107 cells/mL by employing a perfusion culture system with centrifugation and medium exchange in shake flasks and perfusion device in bioreactor. Furthermore, we utilized a design of experiments strategy to systematically modulate a series of transfection-related variables including the quantity of plasmid DNA, the DNA-to-polyethylenimine ratio, incubation duration, and the impact of post-transfection feeding strategies on the yield of recombinant AAV (rAAV). Our comprehensive analysis and subsequent optimizations actualized a remarkable unit yield reaching nearly 2 × 1012 vector genomes (vg)/mL. Importantly, the resulting single-cell yield and biological activity were found to be comparable with those obtained from fed-batch cultures, underscoring the efficacy of our approach. Based on these findings, we investigated rAAV yield via high-density suspend culture in bioreactor, particularly focusing on cell aggregation and the use of perfusion technology. Intriguingly, we attempted to elevate the yield of an oversized recombinant coagulation factor VIII AAV843 vector by 3.5-fold, reaching a yield of 1 × 1012 vg/mL. Concurrently, the medium usage rate was only double that of batch feeding, thereby significantly shrinking the upstream cost of rAAV manufacture. In summary, this strategy significantly benefits large-scale AAV production for both commercial and clinical applications.
Collapse
Affiliation(s)
- Zhe Deng
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Yan-Ling Lv
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Xin-Tao Wang
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Long-Hui Yuan
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Kai Zhao
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Zeng-Min Du
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
| | - Xiao Xiao
- School of Bioengineering, East China University of Science and Technology, Shanghai, China
- Process Development Department, Belief BioMed Co., Ltd, Shanghai, China
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Klimpel M, Pflüger‐Müller B, Cascallana MA, Schwingal S, Lal NI, Noll T, Pirzas V, Laux H. Perfusion Process Intensification for Lentivirus Production Using a Novel Scale-Down Model. Biotechnol Bioeng 2025; 122:344-360. [PMID: 39535315 PMCID: PMC11718438 DOI: 10.1002/bit.28880] [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: 03/23/2024] [Revised: 08/09/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Process intensification has become an important strategy to lower production costs and increase manufacturing capacities for biopharmaceutical products. In particular for the production of viral vectors like lentiviruses (LVs), the transition from (fed-)batch to perfusion processes is a key strategy to meet the increasing demands for cell and gene therapy applications. However, perfusion processes are associated with higher medium consumption. Therefore, it is necessary to develop appropriate small-scale models to reduce development costs. In this work, we present the use of the acoustic wave separation technology in combination with the Ambr 250 high throughput bioreactor system for intensified perfusion process development using stable LV producer cells. The intensified perfusion process developed in the Ambr 250 model, performed at a harvest rate of 3 vessel volumes per day (VVD) and high cell densities, resulted in a 1.4-fold higher cell-specific functional virus yield and 2.8-fold higher volumetric virus yield compared to the control process at a harvest rate of 1 VVD. The findings were verified at bench scale after optimizing the bioreactor set-up, resulting in a 1.4-fold higher cell-specific functional virus yield and 3.1-fold higher volumetric virus yield.
Collapse
Affiliation(s)
| | | | | | - Sarah Schwingal
- Biopharmaceutical Product DevelopmentCSL Innovation GmbHMarburgGermany
| | - Nikki Indresh Lal
- Biopharmaceutical Product DevelopmentCSL Innovation GmbHMarburgGermany
| | - Thomas Noll
- Center for Biotechnology (CeBiTec)University of BielefeldBielefeldGermany
| | - Vicky Pirzas
- Biopharmaceutical Product DevelopmentCSL Innovation GmbHMarburgGermany
| | - Holger Laux
- Biopharmaceutical Product DevelopmentCSL Innovation GmbHMarburgGermany
| |
Collapse
|
9
|
Nascimento A, Faria TQ, Bollmann F, Noverraz M, Pressac G, Roldão A, Peixoto C, Silva RJS. Purification of AAV8 through a scalable two-step monolithic chromatography approach. J Chromatogr A 2025; 1740:465586. [PMID: 39700611 DOI: 10.1016/j.chroma.2024.465586] [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: 08/23/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
Adeno-associated viruses (AAV) are becoming increasingly popular as a powerful tool for gene delivery therapy applications. Although processes to produce AAV are established, future demand for this type of viral vector requires further development of manufacturing processes to make them more robust, scalable, and flexible to accommodate the rise of engineered capsids. This study focuses on designing and evaluating a two-step chromatography process for capturing and polishing AAV8 using monolith chromatography media. A cation-exchange-based capture step was established, using CIMmultus® SO3, resulting in a virus recovery of approximately 70 % of total capsids. This step also achieved high protein removal (>95 %) and considerable DNA clearance (>80 %). For the polishing step, three different CIMmultus® monoliths were evaluated: anion-exchange-based QA (strong anion exchange), multimodal-based PrimaS (weak anion exchange and hydrogen bonding) and multimodal-based PrimaT (weak anion exchange, hydrogen bonding and metal affinity) ligands. High viral vector genome recoveries, DNA and protein clearance and a three-fold full capsid enrichment were observed at a 1 mL column scale. Similar results were obtained during a scale-up to 8 mL and 4 mL monolith volumes for capture and polishing, respectively. These results provide a strong and robust alternative to conventional AAV purification methods, such as resin-based affinity chromatography and density gradient ultracentrifugation.
Collapse
Affiliation(s)
- André Nascimento
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12 2780-901, Oeiras, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Tiago Q Faria
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12 2780-901, Oeiras, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | - Marc Noverraz
- Sartorius Stedim Switzerland AG, 8317 Tagelswangen, Switzerland
| | | | - António Roldão
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12 2780-901, Oeiras, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Cristina Peixoto
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12 2780-901, Oeiras, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ricardo J S Silva
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12 2780-901, Oeiras, Portugal; ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
10
|
Sorroza-Martinez L, Pelletier M, Guay D, Gaillet B. Recent Advances in Therapeutics and Manufacturing Processes of Recombinant Adeno-Associated Virus for the Treatment of Lung Diseases. Curr Gene Ther 2025; 25:237-256. [PMID: 39225214 DOI: 10.2174/0115665232294935240826061311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/01/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Developing delivery vectors capable of transducing genetic material across the lung epithelia and mucus barrier is a major challenge and of great interest to enable gene therapies to treat pulmonary diseases. Recombinant Adeno-associated Viruses (rAAVs) have emerged as attractive candidates among viral and non-viral vectors due to their broad tissue tropism, ability to transduce dividing and quiescent cells, and their safety profile in current human applications. While rAAVs have demonstrated safety in earlier clinical trials for lung disease applications, there are still some limitations regarding rAAV-transgene delivery in pulmonary cells. Thus, further improvements in rAAV engineering are needed to enhance the effectiveness of rAAV-based therapies for lung diseases. Such therapies could benefit patients with chronic lung diseases, such as asthma, chronic obstructive pulmonary disease, pulmonary hypertension, and cystic fibrosis, among others, by regulating hereditary gene mutations or acquired gene deregulations causing these conditions. Alongside therapeutic development, advances in the rAAV production process are essential to meet increasing production demands, while reducing manufacturing costs. This review discusses current challenges and recent advances in the field of rAAV engineering and manufacturing to encourage the clinical development of new pulmonary gene therapy treatments.
Collapse
Affiliation(s)
- Luis Sorroza-Martinez
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - Mia Pelletier
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - David Guay
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
- Feldan Therapeutics, 2666 Boulevard du Parc Technologique Suite 290, Québec, QC, G1P 4S6, Canada
| | - Bruno Gaillet
- Département de génie chimique, Université Laval, Room #3570, 1065 avenue de la Médecine, Pavillon Adrien- Pouliot, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
11
|
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
|
12
|
Shih FH, Chang HH, Wang YC. Utilizing adeno-associated virus as a vector in treating genetic disorders or human cancers. IUBMB Life 2024; 76:1000-1010. [PMID: 38970351 DOI: 10.1002/iub.2896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 06/17/2024] [Indexed: 07/08/2024]
Abstract
Clinical data from over two decades, involving more than 3000 treated patients, demonstrate that adeno-associated virus (AAV) gene therapy is a safe, effective, and well-tolerated therapeutic method. Clinical trials using AAV-mediated gene delivery to accessible tissues have led to successful treatments for numerous monogenic disorders and advancements in tissue engineering. Although the US Food and Drug Administration (FDA) has approved AAV for clinical use, systemic administration remains a significant challenge. In this review, we delve into AAV biology, focusing on current manufacturing technologies and transgene engineering strategies. We examine the use of AAVs in ongoing clinical trials for ocular, neurological, and hematological disorders, as well as cancers. By discussing recent advancements and current challenges in the field, we aim to provide valuable insights for researchers and clinicians navigating the evolving landscape of AAV-based gene therapy.
Collapse
Affiliation(s)
- Fu-Hsuan Shih
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hsiung-Hao Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Yi-Ching Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| |
Collapse
|
13
|
Hsiung KC, Chiang HJ, Reinig S, Shih SR. Vaccine Strategies Against RNA Viruses: Current Advances and Future Directions. Vaccines (Basel) 2024; 12:1345. [PMID: 39772007 PMCID: PMC11679499 DOI: 10.3390/vaccines12121345] [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: 09/29/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The development of vaccines against RNA viruses has undergone a rapid evolution in recent years, particularly driven by the COVID-19 pandemic. This review examines the key roles that RNA viruses, with their high mutation rates and zoonotic potential, play in fostering vaccine innovation. We also discuss both traditional and modern vaccine platforms and the impact of new technologies, such as artificial intelligence, on optimizing immunization strategies. This review evaluates various vaccine platforms, ranging from traditional approaches (inactivated and live-attenuated vaccines) to modern technologies (subunit vaccines, viral and bacterial vectors, nucleic acid vaccines such as mRNA and DNA, and phage-like particle vaccines). To illustrate these platforms' practical applications, we present case studies of vaccines developed for RNA viruses such as SARS-CoV-2, influenza, Zika, and dengue. Additionally, we assess the role of artificial intelligence in predicting viral mutations and enhancing vaccine design. The case studies underscore the successful application of RNA-based vaccines, particularly in the fight against COVID-19, which has saved millions of lives. Current clinical trials for influenza, Zika, and dengue vaccines continue to show promise, highlighting the growing efficacy and adaptability of these platforms. Furthermore, artificial intelligence is driving improvements in vaccine candidate optimization and providing predictive models for viral evolution, enhancing our ability to respond to future outbreaks. Advances in vaccine technology, such as the success of mRNA vaccines against SARS-CoV-2, highlight the potential of nucleic acid platforms in combating RNA viruses. Ongoing trials for influenza, Zika, and dengue demonstrate platform adaptability, while artificial intelligence enhances vaccine design by predicting viral mutations. Integrating these innovations with the One Health approach, which unites human, animal, and environmental health, is essential for strengthening global preparedness against future RNA virus threats.
Collapse
Affiliation(s)
- Kuei-Ching Hsiung
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Huan-Jung Chiang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Sebastian Reinig
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food & Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science & Technology, Taoyuan 33303, Taiwan
| |
Collapse
|
14
|
Roach MK, Wirz P, Rouse J, Schorzman A, Beard CW, Scott D. Production of recombinant adeno-associated virus 5 using a novel self-attenuating adenovirus production platform. Mol Ther Methods Clin Dev 2024; 32:101320. [PMID: 39282074 PMCID: PMC11399549 DOI: 10.1016/j.omtm.2024.101320] [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: 05/23/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024]
Abstract
Recombinant adeno-associated virus (rAAV) has become a prominent vector for clinical use. Despite an increase in successful clinical outcomes, the amount of high-quality rAAVs required for clinical trials and eventual commercial demand is difficult to produce, especially for genetic diseases that are prevalent or require high doses. Many groups are focused on establishing production processes that can produce sufficient rAAV while maintaining potency and quality. Our group used a novel production platform to increase our yield of rAAV5. This production platform uses tetracycline-enabled self-silencing adenovirus (TESSA) to deliver the wild-type AAV replication and capsid genes alongside the adenovirus helper genes necessary for production. Here, we describe our efforts to evaluate the TESSA platform in house. We conducted numerous experiments to determine the optimal conditions for producing rAAV5 from the TESSA production system. We then produced rAAV5 from the TESSA system to compare against rAAV5 produced from triple transfection. Ultimately, we generated data that showed that the vector genome yield of rAAV5 produced with TESSA was >20-fold higher than rAAV5 produced with triple transfection. Additionally, our data show that quality as well as potency in mice of rAAV5 produced with the TESSA system and by triple transfection are equivalent.
Collapse
Affiliation(s)
| | - Phillip Wirz
- BridgeBio Gene Therapy LLC, Raleigh, NC 27607, USA
| | - Jeremy Rouse
- BridgeBio Gene Therapy LLC, Raleigh, NC 27607, USA
| | | | | | - David Scott
- BridgeBio Gene Therapy LLC, Raleigh, NC 27607, USA
| |
Collapse
|
15
|
Xue W, Fulco C, Sha S, Alden N, Panteli J, Hossler P, Warren J. Adeno-associated virus perfusion enhanced expression: A commercially scalable, high titer, high quality producer cell line process. Mol Ther Methods Clin Dev 2024; 32:101266. [PMID: 38868441 PMCID: PMC11166877 DOI: 10.1016/j.omtm.2024.101266] [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: 12/19/2023] [Accepted: 05/10/2024] [Indexed: 06/14/2024]
Abstract
With safety and efficacy demonstrated over hundreds of clinical trials in the last 30 years, along with at least six recent global marketing authorizations achieved since 2017, recombinant adeno-associated viruses (rAAVs) have been established as the leading therapeutic gene transfer vector for rare, monogenic diseases. Significant advances in manufacturing technology have been made in the last few decades to address challenges with GMP production of rAAV products, although yield, cost, scalability, and quality remain a challenge. With transient transfection processes established as a manufacturing platform for multiple commercial AAV products, there remains significant yield, cost, robustness, and scalability constraints that need to be resolved to enable a reliable supply of rAAV products for global patient access. The development of stable producer cell lines for rAAV products has enabled scalability and, in some cases, improvements in productivity. Herein we describe a novel AAV perfusion-enhanced expression (APEX) process, resulting in higher maximum cell densities in the production bioreactor with a 3- to 6-fold increase in volumetric productivity. This process has been successfully demonstrated across multiple serotypes in large scale cell culture with titers approaching 1 × 1012 GC/mL. The APEX production platform marks a significant leap forward in the efficient and effective manufacturing of rAAV vector products.
Collapse
Affiliation(s)
- Wei Xue
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Cameron Fulco
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Sha Sha
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Nick Alden
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Jan Panteli
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - Patrick Hossler
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| | - James Warren
- Ultragenyx Pharmaceutical Inc., Global CMC Development, 19 Presidential Way, Woburn, MA 01801, USA
| |
Collapse
|
16
|
Coplan L, Zhang Z, Ragone N, Reeves J, Rodriguez A, Shevade A, Bak H, Tustian AD. High-yield recombinant adeno-associated viral vector production by multivariate optimization of bioprocess and transfection conditions. Biotechnol Prog 2024; 40:e3445. [PMID: 38450973 DOI: 10.1002/btpr.3445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 03/08/2024]
Abstract
Recombinant adeno-associated viral vectors (rAAVs) are one of the most used vehicles for gene therapy, with five rAAV therapeutics commercially approved by the FDA. To improve product yield, we optimized the suspension production process of rAAV8 vectors carrying a proprietary transgene using a commercially available transfection reagent, FectoVIR-AAV. Using a miniaturized automated 250 mL scale bioreactor system, we generated models of vector genome (vg) titer, capsid (cp) titer, and Vg:Cp percentage from two multivariate design of experiment studies, one centered around bioreactor operating parameters, and another based on the transfection conditions. Using the optimized process returned from these models, the vector genome titer from the bioreactor was improved to beyond 1 × 1012 vg/mL. Five critical parameters were identified that had large effects on the pre-purification vector quantity-the transfection pH, production pH, complexation time, viable cell density at transfection, and transfection reagent to DNA ratio. The optimized process was further assessed for its performance extending to six AAV serotypes, namely AAV1, AAV2, AAV5, AAV6, AAV8, and AAV9 carrying a transgene encoding for green fluorescent protein (GFP). Five of the six serotypes returned higher vector genome titers than the control condition. These data suggest that the choice of transfection reagent is a major factor in improving vector yield. The multivariate design of experiment approach is a powerful way to optimize production processes, and the optimized process from one AAV vector can to some extent be generalized to other serotypes and transgenes to accelerate development timelines of new programs.
Collapse
Affiliation(s)
- Louis Coplan
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, Maryland, USA
| | - Zhe Zhang
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Nicole Ragone
- Research Operations, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - John Reeves
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Audrey Rodriguez
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Aishwarya Shevade
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Hanne Bak
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | - Andrew D Tustian
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| |
Collapse
|
17
|
Crowley L, Cashen P, Noverraz M, Lobedann M, Nestola P. Reviewing the process intensification landscape through the introduction of a novel, multitiered classification for downstream processing. Biotechnol Bioeng 2024; 121:877-893. [PMID: 38214109 DOI: 10.1002/bit.28641] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024]
Abstract
A demand for process intensification in biomanufacturing has increased over the past decade due to the ever-expanding market for biopharmaceuticals. This is largely driven by factors such as a surge in biosimilars as patents expire, an aging population, and a rise in chronic diseases. With these market demands, pressure upon biomanufacturers to produce quality products with rapid turnaround escalates proportionally. Process intensification in biomanufacturing has been well received and accepted across industry based on the demonstration of its benefits of improved productivity and efficiency, while also reducing the cost of goods. However, while these benefits have been shown empirically, the challenges of adopting process intensification into industry remain, from smaller independent start-up to big pharma. Traditionally, moving from batch to a process intensification scheme has been viewed as an "all or nothing" approach involving continuous bioprocessing, in which the factors of complexity and significant capital costs hinder its adoption. In addition, the literature is crowded with a variety of terms used to describe process intensification (continuous, periodic counter-current, connected, intensified, steady-state, etc.). Often, these terms are used inappropriately or as synonyms, which generates confusion in the field. Through a detailed review of current state-of-the-art systems, consumables, and process intensification case studies, we herein propose a defined approach in the implementation of downstream process intensification through a standardized nomenclature and viewing it as distinct independent levels. These can function separately as intensified single-unit operations or be built upon by integration with other process steps allowing for simple, incremental, cost-effective implementation of process intensification in the manufacturing of biopharmaceuticals.
Collapse
Affiliation(s)
- Louis Crowley
- Sartorius Stedim North America Inc, Bohemia, New York, USA
| | - Paul Cashen
- Sartorius Stedim Biotech GmbH, Goettingen, Germany
| | | | | | | |
Collapse
|
18
|
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
|
19
|
Silva CAT, Kamen AA, Henry O. Intensified Influenza Virus Production in Suspension HEK293SF Cell Cultures Operated in Fed-Batch or Perfusion with Continuous Harvest. Vaccines (Basel) 2023; 11:1819. [PMID: 38140223 PMCID: PMC10747379 DOI: 10.3390/vaccines11121819] [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: 10/24/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Major efforts in the intensification of cell culture-based viral vaccine manufacturing focus on the development of high-cell-density (HCD) processes, often operated in perfusion. While perfusion operations allow for higher viable cell densities and volumetric productivities, the high perfusion rates (PR) normally adopted-typically between 2 and 4 vessel volumes per day (VVD)-dramatically increase media consumption, resulting in a higher burden on the cell retention device and raising challenges for the handling and disposal of high volumes of media. In this study, we explore high inoculum fed-batch (HIFB) and low-PR perfusion operations to intensify a cell culture-based process for influenza virus production while minimizing media consumption. To reduce product retention time in the bioreactor, produced viral particles were continuously harvested using a tangential flow depth filtration (TFDF) system as a cell retention device and harvest unit. The feeding strategies developed-a hybrid fed-batch with continuous harvest and a low-PR perfusion-allowed for infections in the range of 8-10 × 106 cells/mL while maintaining cell-specific productivity comparable to the batch control, resulting in a global increase in the process productivity. Overall, our work demonstrates that feeding strategies that minimize media consumption are suitable for large-scale influenza vaccine production.
Collapse
Affiliation(s)
- Cristina A. T. Silva
- Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada;
| | - Amine A. Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada;
| | - Olivier Henry
- Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
20
|
Kilgore R, Minzoni A, Shastry S, Smith W, Barbieri E, Wu Y, LeBarre JP, Chu W, O'Brien J, Menegatti S. The downstream bioprocess toolbox for therapeutic viral vectors. J Chromatogr A 2023; 1709:464337. [PMID: 37722177 DOI: 10.1016/j.chroma.2023.464337] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/20/2023]
Abstract
Viral vectors are poised to acquire a prominent position in modern medicine and biotechnology owing to their role as delivery agents for gene therapies, oncolytic agents, vaccine platforms, and a gateway to engineer cell therapies as well as plants and animals for sustainable agriculture. The success of viral vectors will critically depend on the availability of flexible and affordable biomanufacturing strategies that can meet the growing demand by clinics and biotech companies worldwide. In this context, a key role will be played by downstream process technology: while initially adapted from protein purification media, the purification toolbox for viral vectors is currently undergoing a rapid expansion to fit the unique biomolecular characteristics of these products. Innovation efforts are articulated on two fronts, namely (i) the discovery of affinity ligands that target adeno-associated virus, lentivirus, adenovirus, etc.; (ii) the development of adsorbents with innovative morphologies, such as membranes and 3D printed monoliths, that fit the size of viral vectors. Complementing these efforts are the design of novel process layouts that capitalize on novel ligands and adsorbents to ensure high yield and purity of the product while safeguarding its therapeutic efficacy and safety; and a growing panel of analytical methods that monitor the complex array of critical quality attributes of viral vectors and correlate them to the purification strategies. To help explore this complex and evolving environment, this study presents a comprehensive overview of the downstream bioprocess toolbox for viral vectors established in the last decade, and discusses present efforts and future directions contributing to the success of this promising class of biological medicines.
Collapse
Affiliation(s)
- Ryan Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States.
| | - Arianna Minzoni
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Shriarjun Shastry
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695, United States
| | - Will Smith
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Eduardo Barbieri
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Yuxuan Wu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Jacob P LeBarre
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Juliana O'Brien
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, United States
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695, United States; North Carolina Viral Vector Initiative in Research and Learning, North Carolina State University, Raleigh, NC 27695, United States
| |
Collapse
|
21
|
Leon-Astudillo C, Trivedi PD, Sun RC, Gentry MS, Fuller DD, Byrne BJ, Corti M. Current avenues of gene therapy in Pompe disease. Curr Opin Neurol 2023; 36:464-473. [PMID: 37639402 PMCID: PMC10911405 DOI: 10.1097/wco.0000000000001187] [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] [Indexed: 08/31/2023]
Abstract
PURPOSE OF REVIEW Pompe disease is a rare, inherited, devastating condition that causes progressive weakness, cardiomyopathy and neuromotor disease due to the accumulation of glycogen in striated and smooth muscle, as well as neurons. While enzyme replacement therapy has dramatically changed the outcome of patients with the disease, this strategy has several limitations. Gene therapy in Pompe disease constitutes an attractive approach due to the multisystem aspects of the disease and need to address the central nervous system manifestations. This review highlights the recent work in this field, including methods, progress, shortcomings, and future directions. RECENT FINDINGS Recombinant adeno-associated virus (rAAV) and lentiviral vectors (LV) are well studied platforms for gene therapy in Pompe disease. These products can be further adapted for safe and efficient administration with concomitant immunosuppression, with the modification of specific receptors or codon optimization. rAAV has been studied in multiple clinical trials demonstrating safety and tolerability. SUMMARY Gene therapy for the treatment of patients with Pompe disease is feasible and offers an opportunity to fully correct the principal pathology leading to cellular glycogen accumulation. Further work is needed to overcome the limitations related to vector production, immunologic reactions and redosing.
Collapse
Affiliation(s)
- Carmen Leon-Astudillo
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Prasad D Trivedi
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ramon C Sun
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville FL, United States
- Lafora Epilepsy Cure Initiative, United States
| | - Matthew S Gentry
- Department of Biochemistry & Molecular Biology, University of Florida College of Medicine, Gainesville FL, United States
- Lafora Epilepsy Cure Initiative, United States
| | | | - Barry J Byrne
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Manuela Corti
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
22
|
Moço PD, Xu X, Silva CAT, Kamen AA. Production of adeno-associated viral vector serotype 6 by triple transfection of suspension HEK293 cells at higher cell densities. Biotechnol J 2023; 18:e2300051. [PMID: 37337925 DOI: 10.1002/biot.202300051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/21/2023]
Abstract
In recent years, the use of adeno-associated viruses (AAVs) as vectors for gene and cell therapy has increased, leading to a rise in the amount of AAV vectors required during pre-clinical and clinical trials. AAV serotype 6 (AAV6) has been found to be efficient in transducing different cell types and has been successfully used in gene and cell therapy protocols. However, the number of vectors required to effectively deliver the transgene to one single cell has been estimated at 106 viral genomes (VG), making large-scale production of AAV6 necessary. Suspension cell-based platforms are currently limited to low cell density productions due to the widely reported cell density effect (CDE), which results in diminished production at high cell densities and decreased cell-specific productivity. This limitation hinders the potential of the suspension cell-based production process to increase yields. In this study, we investigated the improvement of the production of AAV6 at higher cell densities by transiently transfecting HEK293SF cells. The results showed that when the plasmid DNA was provided on a cell basis, the production could be carried out at medium cell density (MCD, 4 × 106 cells mL-1 ) resulting in titers above 1010 VG mL-1 . No detrimental effects on cell-specific virus yield or cell-specific functional titer were observed at MCD production. Furthermore, while medium supplementation alleviated the CDE in terms of VG/cell at high cell density (HCD, 10 × 106 cells mL-1 ) productions, the cell-specific functional titer was not maintained, and further studies are necessary to understand the observed limitations for AAV production in HCD processes. The MCD production method reported here lays the foundation for large-scale process operations, potentially solving the current vector shortage in AAV manufacturing.
Collapse
Affiliation(s)
- Pablo D Moço
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Xingge Xu
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Cristina A T Silva
- Department of Bioengineering, McGill University, Montreal, Canada
- Department of Chemical Engineering, Polytechnique Montréal, Montreal, Canada
| | - Amine A Kamen
- Department of Bioengineering, McGill University, Montreal, Canada
| |
Collapse
|